J Stroke Search

CLOSE


J Stroke > Volume 26(2); 2024 > Article
Huang, Lin, and Tzeng: Leukoaraiosis: Epidemiology, Imaging, Risk Factors, and Management of Age-Related Cerebral White Matter Hyperintensities

Abstract

Leukoaraiosis (LA) manifests as cerebral white matter hyperintensities on T2-weighted magnetic resonance imaging scans and corresponds to white matter lesions or abnormalities in brain tissue. Clinically, it is generally detected in the early 40s and is highly prevalent globally in individuals aged >60 years. From the imaging perspective, LA can present as several heterogeneous forms, including punctate and patchy lesions in deep or subcortical white matter; lesions with periventricular caps, a pencil-thin lining, and smooth halo; as well as irregular lesions, which are not always benign. Given its potential of having deleterious effects on normal brain function and the resulting increase in public health burden, considerable effort has been focused on investigating the associations between various risk factors and LA risk, and developing its associated clinical interventions. However, study results have been inconsistent, most likely due to potential differences in study designs, neuroimaging methods, and sample sizes as well as the inherent neuroimaging heterogeneity and multi-factorial nature of LA. In this article, we provided an overview of LA and summarized the current knowledge regarding its epidemiology, neuroimaging classification, pathological characteristics, risk factors, and potential intervention strategies.

Introduction

The term “leukoaraiosis” is derived from the Greek terms “leuko” and “araiosis,” which mean “white” and “rarefaction,” respectively [1-3]. It was originally introduced by Hachinski and his colleagues [3] in 1986 to describe cerebral white matter abnormalities observable as signal hypodensities on computed tomography (CT) scans or hyperintensities on T2-weighted magnetic resonance imaging (MRI) scans [1-3]. Thus, leukoaraiosis (LA) is often referred to simply as white matter hyperintensities (WMHs) or white matter lesions (WMLs) with reference to the cerebral white matter changes.
LA generally manifests early in the fourth decade of life and becomes increasingly more common from age 50 onwards [4-6]. It is also common in healthy elderly individuals [4,7], and most individuals with LA remain asymptomatic. However, LA does not always represent benign imaging features, and could affect normal cognitive ability, motor function, and psychiatric behaviors if the WMLs expand to certain significant regions of the brain [8-13]. Clinically, LA has consistently been reported to be the most common radiological hallmark of cerebral small-vessel diseases (CSVDs) and as being strongly correlated with increased risks of cognitive function decline [14-17], motor gait dysfunction [10,18-20], stroke [21-26], dementia [16,17,21,25], depression [27-30], and even death [21,25,26], Given the clinical importance of WMHs in geriatric populations, there is an urgent need to understand the clinical features and pathogenesis of LA and develop effective strategies for its prevention and management.
In this article, we summarize the current knowledge regarding various aspects of LA—specifically, clinical imaging, epidemiological frequency, pathological characteristics, risk factors, genetic variants and biomarkers, as well as prevention and treatment.

Epidemiology of LA

LA has a high prevalence globally among middle-aged and elderly individuals (36.5%-100% over age 40 years) [4,31-37]. Based on community-based studies in Australia and European countries (including Italy, Spain, France, Netherlands, UK, Sweden, Austria, Germany, and Poland), the incidence of LA in the general population can be as high as 80.0% and 92%, respectively [31,32]. Population-based studies in the Netherlands and Australia have also revealed a high prevalence (up to 95% and 100% in individuals aged >60 years, respectively) [33,34]. Similarly, the incidence of LA in the community-based population in the USA is also high, with the Cardiovascular Health Study and Atherosclerosis Risk in Communities Study reporting an incidence of 85.4%-95.6% in participants aged >55 years (Figure 1) [4,35]; moreover, its reported prevalence is higher among European-Americans (90.2%) than among African-Americans (80.6%) [35]. Recently, the largest community-based study in Asian cities (including Hong Kong, Singapore, and Seoul) reported the prevalence of LA as being up to 85.6% among healthy subjects aged 60-89 years [36] (45.2% in Korea, 80.0% in China, and 97.0% in Singapore) [36] (Figure 1). Community- and hospital-based studies in China have shown that the frequency of LA varied from 36.5%-92.3% among individuals aged >40 years [5,32,37-39] (up to 36.5% in Henan [37], 58.3% in Fujian [5], 73.3% in Beijing [38], and 92.3% in Tibet [39], respectively (Figure 1). Among participants aged >60 years, LA was most frequently reported in individuals from Tibet (98.8%) [39], followed by those from Beijing (90.1%) [38], Shanghai (84.9%) [32], Fujian (67.0%) [5], and Henan (62.7%) [37]. Thus, LA is highly prevalent in elderly populations all over the world and is increasingly becoming a significant public health burden with the rapid increase in the aging population.

Clinical imaging assessment and pathology of LA

LA was first described as abnormal cerebral white matter CT findings visible as hypodensity signals on CT images [1,2,40]. However, compared to CT, MRI is more sensitive in detecting small lesions at an early stage and has therefore become the main clinical imaging diagnostic tool [41]. It is now well-established that LA lesions appear as hypointense WMLs on T1-weighted MRI and hyperintense lesions on T2-weighted MRI [42,43]. Fluid-attenuated inversion recovery (FLAIR) imaging is a form of heavily T2-weighted MRI with the advantage of cerebrospinal fluid (CSF) signal suppression (Figure 2) [44]. Compared to T2-weighted MRI, which simultaneously enhances WML and CSF signals and represents both as hyperintensities [45], FLAIR-MRI can efficiently differentiate WMLs from enlarged perivascular spaces that contain CSF (e.g., Virchow-Robin spaces), as they appear as hypointense regions on FLAIR-MRI. Thus, it greatly improves the detection efficiency of lesions adjacent to CSF-containing spaces and clearly distinguishes small periventricular WMLs (PVWMLs) from the ventricles [45-48]. Currently, FLAIR-MRI probably represents the best imaging method for assessing LA severity.
Several other advanced imaging methods are used for assessing white matter changes [2,48,49]. For example, diffusion tensor imaging (DTI) can not only assess the extent of white matter damage, but also provide information about the integrity of white matter tracts through apparent diffusion coefficient (ADC) or fractional anisotropy (FA) measurements of tissues, which reflect the directionality and rate of water mobility [50,51]. Altered microstructures both within WMLs and in normal-appearing white matter can be identified as regions of elevated ADC and decreased FA on DTI scans [52]. Therefore, DTI can be used to track early white matter changes over time and can provide insights into the in vivo pathogenesis of LA. T2-weighted gradient-recalled echo sequences and corresponding alternative-susceptibility weighted sequences with higher signal and spatial resolution are sensitive to iron deposition [53,54] and can therefore distinguish cerebral microbleeds as small round or ovoid hypointense lesions, which are features of hypertensive CSVD (such as cerebral amyloid angiopathy) as yet undetectable using conventional MRI [55,56].
Clinically, LA can have considerable heterogeneity in terms of its presentation, with various lesion patterns, pathological characteristics, and severities. Originally, LA was classified into two categories based on the spatial locations of WMLs: (1) periventricular LA (with PVWMLs/periventricular WMHs [PVWMHs]), and (2) deep and subcortical LA (with deep WMLs/WMHs [DWMLs/DWMHs]) (Figure 2) [57]. In the former, the lesions are contiguously adjacent to the lateral ventricles; they either appear as irregular PVWMLs or have caps around the frontal horns, a pencil-thin lining, and a smooth halo (Figure 2) [58,59]. Conversely, DWMLs are farther away from the ventricles in the deep or subcortical white matter [58,59] and mainly appear as punctate, early-confluent, and confluent DWMLs corresponding to mild to more severe lesions (Figure 2) [58]. Several methods are being used to classify LA severity [41,60-63]. Unlike fully- or semi-automated segmentation-based volumetric quantification methods [60,64-66], visual rating scales such as the Fazekas scale are easier to use without requiring extensive training and expertise [43,57,63]. They also allow a quicker assessment of LA severity and, despite certain limitations, are widely used both in clinical practice and scientific studies to grade LA as “mild,” “moderate,” or “severe” according to PVWML, DWML, or total WML severity [49]. In order to facilitate the study of the different mechanisms of LA occurrence and progression, it can also be classified as type I and type II LA depending on whether the lesions are considered mild or severe, respectively [67,68].
As described above, PVWMLs and DWMLs represent the two regional categories of LA, and both have different functional relevance and histopathologic correlates that are associated with their spatial relationship to the lateral ventricles [58,59,69]. DWMLs are preferably associated with mood disorders such as depression [70,71]. They are generally attributed (especially early- and complete-confluent lesions) to vascular ischemia and are characterized by patchy demyelination with myelin rarefaction [58,59,72]. Punctate, early-confluent, and confluent DWMLs are considered indicative of mild, extensive, and more severe ischemic tissue damage, respectively [58,59]. In contrast, PVWMLs are mainly linked with cognitive impairment and decline [73-75]. However, periventricular lesions with caps, a pencil-thin lining, and smooth halo are more likely to be due to non-ischemic tissue damage [58,59,72]. Histopathologically, they are mainly characterized by extracellular fluid accumulation, ventricular ependyma disruption, and ependymitis granularis representing ependyma loss and astrocytic gliosis [42,58,59,72,76,77]. However, irregular PVWMLs, like DWMLs, have been shown to have an ischemic origin [59,78,79]. They are generally associated with patchy myelin rarefaction and ischemic tissue necrosis around the perivascular spaces [59,80]; furthermore, unlike DWMLs, which are generally associated with microangiopathy, irregular PVWMLs are more likely to be caused by chronic hemodynamic insufficiency due to focal or systemic hypoperfusion [59,80-82].
Thus, LA lesions are mainly classified into PVWMLs (with caps, a thin lining, and smooth halo) and DWMLs (punctate, early-confluent, and confluent lesions) depending on both relative distance from the ventricular surface and lesion size and severity. PVWML caps, lining, and halos, representing patchy ependyma loss and interstitial fluid leakage, could be non-ischemic in nature, whereas irregular PVWMLs and DWMLs, representing patchy demyelination, are more likely to be attributable to different forms of ischemic tissue damage.
Nonetheless, although PVWMLs and DWMLs differ in terms of certain histopathologic correlates and clinical consequences [12,71,75,77,83,84], this dichotomization of LA lacks a pathophysiological or functional basis corresponding to WMLs, as it is mainly based on the continuity rule associated with relative distance from the ventricular surface. In advanced stages of LA, PVWMLs can coalesce with DWMLs, and it is difficult to clearly distinguish between WML types. Considering the limitations regarding the somewhat arbitrary criteria for classifying PVWMLs/DWMLs, poor objectivity resulting from semiquantitative visual rating scales, and the resulting increase in heterogeneity of assessment and reduction in the consistency of findings across studies, a new LA subclassification method based on WMH characteristics was proposed by Kim et al. [59] in 2008. Its subclasses have etiological and functional relevance, which reduces the WML finding heterogeneity between studies. This scheme uses a finer quantification method to classify LA lesions into four classes: (1) juxtaventricular WMLs (JVWMLs): lesions—located in juxtaventricular areas within 3 mm of the ventricular surface, (2) PVWMLs: lesions in the periventricular watershed zone (3-13 mm from the ventricular surface, (3) DWMLs: lesions 13 mm or further from the ventricular surface, and (4) juxtacortical WMLs (JCWMLs): deep lesions located in juxtacortical white matter areas within 4 mm from corticomedullary junction [59]. Of these, only JVWMLs are non-ischemic [58,59,81,82]. They are more likely attributable to CSF leakage into the adjacent brain parenchyma because of their direct attachment to the ventricular surface [59,85-87]. Both PVWMLs and DWMLs are characterized by ischemia-induced disruption of long white matter tracts [58,59,77,86], and might result from hypoperfusion and CSVD, respectively [59,77,84,86]. Like ischemic PVWMLs and DWMLs, JCWMLs also have an ischemic origin [59]; however, they could have a different pathological basis [59] and are characterized by CSVD-associated disruption of U-fibers rather than the disruption of long white matter tracts [59].
Notably, despite the considerable pathological, functional, and neuroimaging heterogeneity in LA lesions, the correlations between them have not been fully understood, and we still need to identify specific WML subclasses and determine the underlying clinical factors and subsequent clinical consequences. Recently, Jung et al. [88] comprehensively quantified the characteristics of WMLs through a novel, fully automated procedure that uses hierarchical clustering methods to classify LA into three distinct classes based on features including lesion contrast, noncontiguous lesion number, volume of each non-contiguous lesion, and periventricular to deep lesion volume ratio. They defined class I LA as the presence of small, punctate, scattered, relatively lower-contrast, and deep WMLs [88]; class II LA as the presence of large, patchy, irregular, or confluent lesions, predominantly in the periventricular white matter; and class III LA as the presence of mild and relatively higher-contrast lesions restricted to the juxtaventricular white matter [88]. The three classes have different pathological features and distinct correlations with clinical factors and/or outcomes. Pathologically, class II LA lesions are characterized by lower myelin content than class I and class III lesions, indicating more serious myelin rarefaction.88 They are more common in older subjects with hypertension and/or lower physical activity levels, whereas class I lesions are more common in subjects with poor sleep quality [88]. Therefore, compared with the previous methods, this fully automated, hierarchical clustering based classification can provide more details on WML features. It can be used to distinguish between LA subclasses with different clinical factors and consequences, and facilitates the understanding of the correlations between specific subclasses of WML burden and the underlying clinical features and pathophysiologies.

Clinical risk factors for LA

As a prevalent age-related manifestation of CSVD in elderly individuals, LA is recognized as being multi-factorial in nature and having many potential risk factors identified by a large number of studies (Figure 3). As some of these findings are inconsistent across different studies, possibly due to differences in study population, methodology, sample size, and participant ethnicity, we will introduce and discuss the most widely studied risk factors below.

Age

Age is the most important risk factor for LA, and LA prevalence increases with age (reported as about 50.9% in the 40s [6], 78.0% in the 50s [35], and 80.0%-95.6% at age ≥60 years in the general population worldwide) [4,31-33,36]. According to a Chinese community-based study, the frequency of periventricular and deep LA increase from only 49.8% and 45.8% in the 40-49 years to 73.5% and 63.5% in the 50-59 years, 87.7% and 83.2% in the 60-69 years, and 97.1% and 89.5% in the 70-79 years age groups, respectively [38]. Another study on hospitalized Chinese patients showed that the frequencies of mild and moderate to severe LA rose from 21.1% and 6.7% in the 40-49 years to 29.2% and 16.2% in the 50-69 years and from 37.4% and 20.0% in the 60-69 years to 41.2% and 40.5% in the ≥80 years age groups, respectively [5].
LA is also known to progress with age, although progression varies in different populations and does not necessarily occur in every case [89-91]. Age has been shown to be a predictor of LA progression in a few longitudinal studies [91-94], and increased age is strongly correlated with increased risk of lesion worsening, especially in low initial grade LA [91]. Older age is also significantly associated with a higher emergence rate and faster progression of LA and higher percentage of change in LA volume [92-94]. However, other longitudinal studies have not supported these findings and instead found baseline WMH burden to be a predictor of LA progression [58,90,95-97]. Among these, two reports from the Austrian Stroke Prevention Study [58,90], which involved 3-year and 6-year follow-ups of healthy community-dwelling individuals, showed that the increase in WML volume in subjects with early confluent and confluent WMLs was significantly more rapid at both follow-up time-points than those in subjects without lesions and with punctate lesions at baseline, suggesting that baseline WML severity is a predictor for LA progression. Similarly, a recent prospective case-control study also found that high WMH burden at baseline was significantly associated with LA progression (odds ratio: ≤7.68) and more subjects in the high baseline WMH group had LA progression at follow-up than in the low baseline WMH group (77.6% vs. 34.7%) [96]. Moreover, baseline WMH burden and LA progression are significantly correlated for both DWMLs and PVWMLs [94,95,98]. The increase in DWML volume is reported to be greater than that in PVWML volume [95,98]; moreover, a recent study reported that PVWML progression frequency is lower than early-confluent and confluent DWML progression frequency at 3-year (22.9% vs. 38.3%) and 6-year follow-ups (42.9% vs. 74.0%) [58]. Taken together, these findings suggest that the major determinant and predictor of LA progression is not age, but baseline WML level. Given the relationships between age and LA prevalence and severity, we think that age could contribute indirectly to LA progression through its influence on baseline WML volume.

Blood pressure and hypertension

Like aging, hypertension is also strongly associated with LA. It can increase both the prevalence and severity of LA and is thus considered an independent and important risk factor for it [35,99-106]. Moreover, both higher diastolic blood pressure (DBP) and systolic blood pressure (SBP) have been shown to be significantly associated with the risk of LA [107-109]. A large meta-analysis of multi-ancestry genome-wide association studies (GWAS) on WML volume has also provided evidence of causative associations between higher DBP and SBP and higher WML volume in participants with and without hypertension [110]. Another recent meta-analysis has also suggested a consistent and strong association between LA severity and DBP and SBP [111]. However, evidence from some independent studies suggests different associations between LA and both DBP and SBP [4,100,107,112-115]. Currently, the risk of LA is considered to be correlated with DBP in mid-life and SBP in later life [108,109], suggesting that DBP control in early mid-life and SBP control later in life could protect against increased risk and severity of LA.
Although some longitudinal studies did not find any significant association between hypertension and LA progression after adjustment for baseline WML burden or other clinical factors [18,90,92,95], hypertension is considered to be correlated with LA progression [5,116-119]. This is because several studies have shown that anti-hypertensive treatment can slow WML volume increase, suggesting a role of hypertension in promoting LA progression [101,120-125]. Recently, a prospective case-control study in an Asian population identified both baseline WML burden and hypertension as important risk factors for LA progression [96]. Furthermore, DBP and SBP have been associated with LA worsening [89,91,94,126]. A recent meta-analysis study including 12 closely related studies on the role of blood pressure (BP) in the progression of WML revealed that both SBP and DBP elevation can promote WML progression, suggesting that both are important risk factors for LA progression [127]. This study also showed that SBP and DBP had different effects on LA progression and that DBP increase had a greater effect on lesion worsening, particularly in patients aged <70 years [127]. Thus, delaying LA progression requires the development of personalized strategies for controlling BP levels.
Taken together, these findings indicate that, like hypertension, high SBP and DBP are associated with not only the incidence and severity of LA, but also its progression. Nevertheless, the precise role of high BP in the pathogenesis of LA remains unclear, although some high BP-mediated mechanisms that could underlie this pathogenesis are as follows: (1) high BP could induce vessel wall thickening and lumen narrowing, subsequently reducing blood flow and leading to ischemia-related tissue damage in the white matter, (2) high BP could cause endothelial damage followed by blood-brain barrier breakdown, resulting in the leakage of potentially toxic substances into the brain and subsequent cell injury, and (3) endothelial damage could induce endothelial inflammation, induce autoimmune reactions against the myelin on axonal fibers, and eventually result in demyelination and even axonal degeneration [49,59,72,86,128,129]. Future in vitro and in vivo studies will be required to confirm which of these proposed mechanisms are involved in LA.

BP variability

Compared to hypertension and absolute BP elevation, BP variability (BPV) generally receives less attention, but has recently been shown to increase the risk of both stroke and dementia [130-132]. Moreover, a growing number of studies have found that BPV is linked to CSVD-related phenomena such as LA, cerebral microbleeds, and lacunes [133-143]. Most of these studies showed that increased SBP variability was related to a higher risk or burden of LA [133,135,136,138,141,143], although a few studies did not find any significant associations [144-146]. A recent meta-analysis of population-based prospective cohort studies also found a significant and positive association between SBP variability and LA [147]. In contrast to SBP variability, the findings regarding the relationship between DBP variability and LA are more conflicting. Most studies found that DBP variability did not affect LA risk or lesion volume [136,138,139,144,145,147]. However, two recent studies have reported a significant association between diastolic BPV and LA [141,146]. One was a longitudinal study that showed that the 24-hour average real variability of DBP, an index of BPV, was associated with LA progression in participants with cardiovascular diseases [146]. In the other study, both increased SBP and DBP variability were associated with increased LA volume, especially PVWMH burden, independently of BP levels [141]; moreover, DBP variability was significantly associated with LA volume [141]. The reasons for the inconsistencies in results may be due to differences between the studies in terms of the BPV indices used and in study populations and the methods of evaluating LA. Thus, results regarding the effects of DBP variability on LA should be interpreted cautiously. Overall, the available evidence strongly suggests that systolic BP fluctuation is a risk factor for LA and that monitoring and controlling BP fluctuations could help prevent LA and improve its prognosis.

Diabetes mellitus

As a vascular risk factor, diabetes mellitus may also contribute to the increased risk of LA. A number of studies have investigated the effect of diabetes mellitus on LA, but their findings are inconsistent, possibly due to differences in sample sizes, participant ethnicities, and statistical methods across studies. Both older [7,148,149] and more recent studies with large sample sizes have shown that diabetes mellitus is significantly associated with LA volume in some European populations [106,125,150]. The largest community-based study (up to 37,041 participants from the UK Biobank cohort) also found a strong correlation between diabetes mellitus and WML volume both before and after adjusting for BP, age, sex, and other cardiovascular risk factors (such as smoking) [109]. Conversely, most studies on Asian populations, except our cross-sectional study on hospitalized Chinese patients [5], failed to confirm any relationship between diabetes mellitus and LA [93,114,151,152]. Additionally, diabetes mellitus was shown to be correlated with LA progression in both studies [5,92], although longitudinal studies in other countries did not find any associations between diabetes mellitus and WML progression [94-96]. Thus, it is likely that diabetes mellitus is not associated with the progression of LA, but is strongly correlated with LA frequency and severity, particularly in European populations. Further studies are required to investigate the associations between diabetes mellitus and LA incidence and progression and explore the effects of antidiabetic treatments on the prevention and management of LA.

Smoking

Like diabetes mellitus, smoking may also be correlated with LA. Although some studies in Asian populations did not find any relationship between smoking and LA [93,114,151,152], many studies in non-Asian populations, including the Cardiovascular Health Study, Rotterdam Scan Study, Atherosclerosis Risk in Communities Study, and Framingham Offspring Cohort Study, found significant associations between smoking and LA [4,33,91,94,99,106,125,148,153-155]. Some of these studies in European and American populations found that smoking was associated with the incidence and severity of LA [99,106,125,148,153,155]. Recently, the largest genetic study on complex CSVD till date revealed a strong causal association between increased lifetime cigarette smoking (specifically, the lifetime smoking index) and higher WML burden in an older community-based population (up to 50,970 individuals from the cohorts for heart and aging research in genomic epidemiology (CHARGE) and from UK Biobank [110]. Another large study (with ten thousand European participants from the UK Biobank cohort) showed that smoking was strongly related with increased WML load both before and after adjustments for BP, age, sex, and other cardiovascular risk factors (such as diabetes mellitus) [109]. Smoking has also been correlated with LA progression. Except two studies with limited sample sizes in Asian and Australian populations [95,96], most studies in European and American populations suggest that cigarette smoking is a risk factor for LA progression [4,33,91,94,99,153-155]. Thus, smoking is likely to be associated with LA incidence and severity as well as its progression, although the possible biological processes through which it could mediate the pathogenesis of LA remain unclear.

Dyslipidemia

The findings of studies on the effects of dyslipidemia on WMLs are also conflicting. One study found a strong inverse association between hyperlipidemia and LA severity [156]; specifically, acute ischemic stroke patients without hyperlipidemia had more severe LA than those with a history of hyperlipidemia [156]. Both hypercholesterolemia and hypertriglyceridemia have been shown to be significantly associated with lower risk and decreased severity of LA [157,158]. This suggests that lipid metabolism-associated factors have a protective effect on LA severity. However, two recent studies have reported that dyslipidemia has deleterious effects on the risk and burden of LA [125,159]. In one study, single modeling of individual vascular risk factors-global brain associations was used to show that hypercholesterolemia was significantly associated with higher WMH load in a UK Biobank cohort [125]. The other study was a meta-analysis that showed that individuals with hyperlipidemia were more likely to have LA than those without hyperlipidemia [159]. These findings suggest that hyperlipidemia is a risk factor for LA; however, several other studies failed to find any associations between hyperlipidemia or hypercholesterolemia and LA [38,39,125,152]. Furthermore, a longitudinal study showed that increased high-density lipoprotein cholesterol (HDL-C) and decreased low-density lipoprotein cholesterol (LDL-C) levels could increase the risk of LA progression [91], although other studies did not find any such associations [38,110,160]. Due to these inconsistent results, it is not clear whether lipid metabolism-associated factors are associated with LA incidence or progression. Future studies are therefore needed to clarify the relationships between dyslipidemia and LA, and caution should be exercised when considering lipid-modifying therapies for patients with LA.

Arterial stiffness

Arterial stiffness is a known predictor of cardiovascular disease and is known to be related to CSVD. Some studies have shown that increased arterial stiffness is associated with higher WMH burden [161-166]. Moreover, a systematic review and meta-analysis found consistent associations between arterial stiffness and CSVD markers, including LA, cerebral microbleeds, and cerebral infarcts, across several cross-sectional studies [167]. The relationships between arterial stiffness and LA have been widely investigated in recent years. Except in one study, which found no association between pulse wave velocity (PWV), considered as the gold standard for measuring arterial stiffness, and cerebral SVD markers [168], most studies have shown that increased arterial stiffness is significantly associated with increased WMH prevalence or volume [169-172]. A few studies have also investigated the relationships between arterial stiffness and the LA subtypes classified on the basis of PVWMH and DWMH, but their results were inconsistent [173-176]. Two of these studies reported that PWV was statistically significantly associated with both LA subtypes [173,174]. However, two other studies found significantly different associations between arterial stiffness and PVWMH and DWMH [175,176]—in one study, arterial stiffness was associated with PVWMH but not DWMH [175], whereas in the other, PVWMH had a higher correlation with arterial stiffness than DWMH [176]. Although the differences between the associations between arterial stiffness and PVWMH versus DWMH need to be clarified, these findings strongly suggest that arterial stiffness is associated with LA incidence and severity.
Arterial stiffness has also been correlated with LA progression. Several longitudinal studies have shown that arterial stiffness is related to LA progression in both community-dwelling older adults and patients with type 2 diabetes [177-179]. They found that baseline PWV was higher in subjects with WMH progression than in those who did not and that the WMH progression rate was higher among individuals with higher PWV [177,178]. Thus, arterial stiffness could be a crucial cause of rapid LA progression in older individuals. Recently, a systematic review and meta-analysis characterized the associations between CSVD markers (such as LA, lacunes, perivascular spaces, cerebral microbleeds, and recent small subcortical infarcts) and cerebrovascular reactivity, cerebral autoregulation, and arterial stiffness [180]. Although the associations between LA and measures of cerebrovascular regulation and arterial stiffness have not been assessed independently, the significant associations between CSVD markers and increased arterial stiffness and impaired cerebrovascular reactivity seem to indicate that cerebrovascular regulation and arterial stiffness may have some role in the development or progression of LA [180].
Collectively, these findings suggest a strong association between arterial stiffness and LA. We believe that arterial stiffness should be considered a risk factor for LA and that it represents a potential mechanism of LA onset and progression.

Homocysteine and vitamin levels

Homocysteine is a naturally occurring sulfur-containing amino acid that can induce oxidative injury, endothelial dysfunction, and vascular damage [181-183] and is known to increase the risk of cardiovascular and cerebrovascular diseases [184-186]. Most cross-sectional studies, except a few [187-189], have consistently shown that total plasma homocysteine (tHcy) level elevation, or hyperhomocysteinemia, is significantly associated with LA [181,190-204]; a few longitudinal studies have also shown that it can increase the risk of LA progression [205,206]. This strongly suggests that hyperhomocysteinemia is a risk factor for LA. Furthermore, the detrimental effects of high homocysteine levels in LA are related to the locations of WMLs and sex-related differences [207-209]. One study showed that high tHcy levels were independently correlated with increased deep LA but not periventricular LA in healthy community-dwelling individuals and that this association was significant only in men [207]. However, other studies found that plasma tHcy levels were more significantly associated with periventricular and frontal LA rather than deep and subcortical LA in both stroke patients and healthy individuals [208,209]. This indicates differences in etiologies between the two main subtypes of LA, and also suggests the possible involvement of dysregulated tHcy metabolism and subsequent endothelial dysfunction in the pathogenesis of PVWMH.
Hyperhomocysteinemia may be attributed to the deficiency of vitamins such as folic acid, vitamin B6, and vitamin B12 [210,211]. These vitamins have been implicated in various cognitive function and vascular disorders, including LA [212-215]. Low plasma vitamin B12 level is significantly associated with more severe LA, especially periventricular LA [216,217]. Moreover, low baseline vitamin B12 level was shown to be significantly associated with PVWMH progression in lacunar stroke patients in one longitudinal study [218], although other studies found no associations with total WMH or WMH progression [206,219-223]. In the only one of these studies with a sub-group analysis on WMH, there was a significant association between deep WMH and vitamin B12 levels in patients with major depression [221]. Due to the inconsistencies in these results, due to multiple possible reasons, more cross-sectional and longitudinal studies will be needed to validate the association between vitamin B12 levels and LA. Nevertheless, animal model studies on myelin morphology suggest that vitamin B12 deficiency has a harmful effect on WMLs characterized by demyelination [224,225]. Overall, the available evidence seems to suggest that low plasma vitamin B12 level is a risk factor for LA, especially with PVWMH.
Like vitamin B, vitamin D, which plays important roles in bone metabolism regulation and cognitive functioning, has also been studied in elderly individuals with LA [226-228]. Some studies have shown that lower serum 25-hydroxyvitamin D levels were negatively correlated with WMH volume, suggesting that vitamin D deficiency is a risk factor for LA [229-236]. Another study revealed a stronger correlation between vitamin D and PVWMH rather than DWMH [237]. However, other studies, including several longitudinal studies, did not find any association between vitamin D levels and WMH [238-242]. These inconsistencies in results could be due to differences in study populations, LA and vitamin D level evaluation methods, LA lesion and etiology heterogeneities, differences in statistical power due to differences in sample sizes, presence of concomitant disorders, and so on. Future longitudinal studies and prospective clinical trials on vitamin D supplementation are required to clarify any possible causal relationships with LA.

Obesity

Obesity is an important risk factor for cardiovascular diseases. It is generally assessed using the body mass index (BMI) and waist-to-hip ratio (WHR), and these measures can also be considered as cardiovascular risk factors. Obesity has been also shown to be associated with LA [160,243,244]—both higher BMI and WHR were shown to be significantly associated with higher WMH burden, suggesting that obesity is a novel risk factor for LA [69,125,245]. Recently, the association between obesity and LA was shown to depend on sex and race [246,247]. In one study, there were significant differences between men and women in terms of the association between BMI and DWMH; moreover, only male sex interacted with higher BMI to result in increased DWMH volume [246]. In another study, obesity was found to significantly increase the risk of WMH in African Americans [247]. These findings indicate that obesity is associated with LA severity and that it could be involved in the pathogenesis of LA. Another recent study showed that visceral obesity contributed to a higher DWMH/PVWMH ratio through elevated levels of the pro-inflammatory cytokine interleukin-6, independent of age and sex, thus providing insights into the possible inflammatory mechanisms underlying DWMLs [245], although future studies are required to confirm this hypothesis at the molecular level and using imaging assessment and animal models.

Education level

Some studies have reported that education levels are associated with LA [70,106,248-251]. In one study, there were significant negative associations between education level and both WMH frequency and volume [106], suggesting a protective effect of a higher education level on the incidence and severity of LA. Recently, a systematic review and meta-analysis including six studies also showed that individuals with low educational levels had more WMHs compared to individuals with higher educational levels [252]. However, other studies did not find any associations between LA and education level [36,192,237,253,254]. Differences in the definitions and assessment of education levels and in the statistical analysis methods used may have contributed to these discrepancies. Therefore, results regarding the relationship of LA with education level should be interpreted cautiously, because it is a complex parameter that can have significant effects on lifestyle and socioeconomic status in later life, which in turn have also been shown to be correlated with LA [4,248,252].

Sex

Many studies have shown that LA incidence and progression tend to be higher among women [4,33,34,39,93,94,98,99,255-257]. However, other studies [91,92,95,258,259], including several recent ones, could not support these findings and found no such differences between women and men [36,96,160,251]. Sex-related differences in the associations between risk factors and LA have also been observed in some studies. For example, the associations between LA and hypertension [136,260], diabetes [149], and atherosclerosis [261] are stronger in men than in women, possibly due to the higher prevalence of vascular risk factors in men. A recent study examined the possible moderating effects of sex on the associations between such risk factors and LA in a large cohort of community-dwelling individuals without dementia and found several differences among women and men [246]. Specifically, sex was found to be significantly associated with total WMH volume independently of age, hypertension, and hip-to-waist ratio (HWR). Moreover, both age and HWR were risk factors for WMH burden in women, whereas multiple risk factors—including age, hypertension, HDL level, HWR, and BMI—were significantly associated with higher WMH volume in men [246]. The differences in associations between BMI and DWMH between men and women strongly suggest that sex moderates the associations between these risk factors and LA and has important effects on LA incidence, severity, and progression. Although the differences in the underlying mechanisms in LA between women and men are not well understood, they could be explained by differences in genetic factors and susceptibility to ischemia and hormonal changes later in life.

Ethnicity

Ethnicity has also been reported to affect the prevalence, severity, and progression of LA [32,35,150,262-264]. African Americans and Mexican Americans have been reported to have higher WMH volumes compared to non-Hispanic whites [150,262]. Additionally, the confluent WMH prevalence and WMH progression rate were higher in African Americans than in European Americans [35,263,264]. Asians have also been reported to have a higher WMH burden than White Australians [32]. However, there were no significant differences in WMH burden (both overall and local WMH burden) between Asians and Europeans [114]. Among Asian regions, the prevalence of moderate-to-severe WMHs was higher in Singapore than in China and Korea [36,152]. Additionally, some studies have reported ethnicity-related differences in the associations between certain risk factors and LA [35,114,150]. For example, the associations between WMH burden and age, DBP, and National Cholesterol Education Programme Adult Treatment Panel III cardiovascular risk scores were stronger among South Asians than among Europeans, whereas the association between WMH burden and diabetes mellitus duration was stronger among Europeans than among South Asians [114]. Recently, a study showed that age could predict high WMH burden only for European Americans and that only obesity could predict high WMH burden among African Americans [247]; it also reported that the deleterious effect of obesity on WMH load was more pronounced among African Americans than European Americans [247]. Together, these findings indicate that ethnicity affects LA risk and modulates the effects of risk factors on LA. Although the mechanisms underlying the ethnicity-related differences in LA risk remain unclear, they could be attributable to differences in vascular risk factor susceptibility, lifestyles, genetic and environmental factors, and susceptibility to developing cardiovascular diseases.

Genetic risk factors for LA

Like various vascular risk factors and sex- and ethnicity-related differences, genetic predispositions can also explain some of the variance in LA risk and burden. LA shows a high heritability and has a strong genetic basis [265-267]. Many LA susceptibility genes have been identified in the past decades through genetic studies with different study designs on different ethnic cohorts [251,268-286]. Of these, candidate gene association studies have identified up to 52 susceptibility genes significantly associated with LA (Figure 4A) [67,251,268-275,277-284]. Two recent exome-wide association studies on individuals of European and African descent and UK Biobank subjects revealed that 10 single nucleotide polymorphisms in eight genes (TRIM65, ACOX1, CARF, FBF1, MRPL38, NBEAL1, WDR12, and GBE1) are significantly associated with the risk of LA (Figure 4A) [270,287]. Moreover, two GWASs on stroke-free European individuals and multi-ethnicity cohorts (including individuals of European, African, Hispanic, and Asian ancestry) free of both stroke and dementia have identified a large number of genetic variants at multiple loci associated with increased LA risk and burden [285,286]. The variants in 13 of these genes (TRIM65, TRIM47, WBP2, FBF1, ACOX1, PDCD11, UNC13D and NEURL on Chr17q25, SH3PXD2A and TAF5 on Chr10q24, EFEMP1 on Chr2q16, HAAO on Chr2p21, and PMF1 on Chr1q22) reached genome-wide significance [270,285,286]. Recently, five meta-analyses of GWASs on WMH burden confirmed the presence of most of these variants in individuals from the UK Biobank and CHARGE consortium cohorts, and further identified up to 63 other susceptibility genes with genome-wide significance, including PLEKHG1, NBEAL1, KLHL24, CARF, WDR21, ICA1L, DEGS2, DCAKD, ECHDC3 and NMT1, and so on (Figure 4B) [110,288-291]. Of the 76 susceptibility genes identified by GWASs, 21 (TRIM65, TRIM47, WBP2, EFEMP1, SH3PXD2A, PLEKHG1, C16orf95, COL4A2, NBEAL1, NMT1, HAAO, ACOX1, UNC13D, FBF1, DEGS2, DCAKD, KLHL24, ICA1L, WDR12, CARF, AC098824.6) are common in at least two GWASs mentioned above (Figure 4B). Furthermore, 13 of these LA susceptibility genes identified by GWASs have been confirmed by candidate gene association studies (TRIM47, WBP2, PMF1, COL4A2, NOS3, APOE) and whole exome sequencing studies (TRIM65, ACOX1, MRPL38, FBF1, WDR12, NBEAL1, CARF) (Figure 4C) and are therefore the most reliable risk genes for LA. It is worth mentioning that one meta-analysis of GWAS considered LA subtypes separately and detected differences in risk genes between PVWMH and DWMH in two multi-ethnicity study cohorts (primarily white, along with black and Hispanic individuals) [291]. It identified 15 specific risk genes for PVWMH (EFEMP1, CARF, ICA1L, KRT8P15, WDR12, AC098824.6, AC023271.1, AC023271.2, AC098831.4 and AC010900.2 on Chr2, COL4A2 on Chr13, PLEKHG1 on Chr6, NOS3 on Chr7, C16orf95 on Chr16, and NMT1 on Chr17); only one risk gene (RP11-137H2.6) was found to be specific to DWMH (Figure 4D-E). Furthermore, PVWMH and DWMH shared 20 risk genes with genome-wide significance, most of which are located at Chr17q25. Of these, nine common genes (TRIM65, TRIM47, WBP2, UNC13D, MRPL38, ACOX1, FBF1, RP11-552F3.9, and RP11-552F3.12) (Figure 4D-E) were identified in both the discovery and replication cohorts.
Studies have also found several genes, other than the previously discussed variant genes, with abnormal mRNA levels in LA patients [292-294]. Two studies used whole-blood gene expression profiling and found up to 184 differentially expressed genes between LA patients and healthy individuals (Figure 4F).293,294 Among those significant genes, nine genes were shown to be dysregulated in WMH lesion tissue (Figure 4F).292,294 Of these, only 7 genes (SLC15A2, PARVA, AHNAK, KLHL6, PIP5K1B, ALAS2, and SEPT11) showed consistent changes between whole blood and brain tissue [292-294]. Other than these genes, 345 other genes also had abnormal mRNA expression in LA lesions compared to that in normal brain tissues (Figure 4F) [292]. These dysregulated genes are functionally associated with immune and cell cycle regulation, apoptosis, proteolysis, ion transport, cell structure, electron transport, and metabolism, suggesting the potential molecular mechanism underlying the pathology of LA [292]. Recently, another whole-blood gene expression study identified 148 dysregulated genes associated with LA progression (30 up-regulated and 118 down-regulated genes) [295]. Among these, two downregulated genes (TTC9 and WLS) are shown to be upregulated in WMH lesions [292,295], reflecting the inconsistent gene regulation between the peripheral and central systems (Figure 4F). According to integrated analyses on this abnormal gene expression, we know that few genes were common across different whole-blood gene expression profiles in LA. This heterogeneity may be explained by differences in disease course (early vs. late stage LA), study populations (European vs. American), study methods (gene microarray vs. transcriptome sequencing), as well as the difference in RNA stability and reliability in samples (whole blood vs. solid tissue). Given the strong heterogeneity of the results described above and the limited gene expression studies on LA tissues, we think that it is necessary to perform a comprehensive cross-omics analyses on LA using WMH lesion tissues from large multiethnic study populations in the future.
In order to further identify gene variants that may contribute to the pathogenesis of LA by influencing expression, stability, and/or function, we performed an integrated analysis of the dysregulated and variant genes involved in LA and identified three variant genes with abnormal expression in LA lesions (two upregulated genes, MS4A6A and TNKS, and one down-regulated gene, EVPL) (Figure 4G). Each of the up-regulated genes only had one variant with GWAS significance in LA. The variants rs144406103 on MS4A6A and rs11249945 on TNKS affect the 3’ untranslated region and an intron of the gene, respectively. Although the effects of the variants on the stability and function of those two up-regulated genes may be limited, both MS4A6A and TNKS could directly participate in the pathology of LA lesions. MS4A6A encodes a member of the membrane-spanning 4A gene family. It is involved in the regulation of soluble TREM2 and is linked to Alzheimer’s disease [296]. TNKS, encodes tankyrase, which has histone binding, pentosyltransferase, and zinc ion binding activities [297]. It has been shown to be involved in the regulation of Wnt/β-catenin signaling and has been implicated in various cancers [297,298]. In contrast, the down-regulated gene EVPL has up to 22 variants significantly associated with LA. These include variants affecting the 3’ untranslated region (rs1128889 and rs1135531) and missense variants (rs2071192 and rs2071193). EVPL encodes a member of the plakin family of proteins that contributes to the formation of desmosomes and the epidermal cornified envelope [299] and has been shown to be associated with oesophageal squamous cell carcinomas [299]; however, its functions are poorly understood.
Taken together, both genetic variations and dysregulated gene expression are important risk factors for LA. As the roles of these variations and dysregulated genes in the etiology of LA have been poorly understood to date, the precise functions of these genes must be explored at the molecular, cellular, and imaging levels.

Management of LA

The imaging changes in LA are irreversible, as they eventually progress and enlarge to affect the surrounding cerebral white matter. Since the pathogenic mechanisms of LA are poorly understood, it is difficult to treat WMLs and reverse their formation. Therefore, what matters most currently is to delay the onset of LA, attenuate its progression, and reduce its incidence and severity through effective strategies. To our knowledge, the current prevention and management methods for LA are mostly empirical and mainly target vascular risk factors to prevent or delay the progression of LA through pharmacological interventions. During the past twenty years, more and more evidence has shown the efficiency of controlling for vascular risk factors in the management of LA. Here, we have reviewed these potential intervention strategies.

BP control

Hypertension is a crucial risk factor for LA, and high SBP and DBP levels are also associated with LA progression. Thus, BP-lowering therapy is considered an effective strategy for LA prevention and management. With the exception of some studies [300-302], most studies support the view that effective BP control can decrease the progression of LA and reduce the risk of severe LA [101,102,117,120,121,123,303]. Multiple cross-sectional studies have shown individuals who were successfully treated using anti-hypertension therapies had a lower risk of LA than those with poorly controlled hypertension [101,102,117,121]. Similarly, several randomized controlled trials (RCTs) including a Perindopril Protection Against Recurrent Stroke Study substudy [120,304], Systolic Blood Pressure Intervention Trial Memory and Cognition in Decreased Hypertension (SPRINT-MIND) substudy [123], and the Three-City (3C)-Dijon MRI study [303], also showed that active BP control can significantly decrease the risk and/or mean total volume of incident LA during the follow-up period. Additionally, one Prevention of Dementia by Intensive Vascular Care trial MRI substudy found that hypertensive intervention was more effective in patients with high baseline WMH burden [305]. These findings suggested the beneficial effects of BP control on preventing LA. Furthermore, the effects of intensive and standard BP-lowering or more conservative treatments have also been compared. Three prospective, randomized clinical studies—a SPRINT-MIND substudy [124], Action to Control Cardiovascular Risk in Diabetes Memory in Diabetes (ACCORD-MIND) substudy [306], and the Intensive Versus Standard Blood Pressure Lowering to Prevent Functional Decline in Older People study [307]—showed that intensive BP control did result in less LA progression compared to standard BP control among hypertensive patients. The results of a recent meta-analysis of randomized trials also support the conclusion that intensive BP-lowering therapy prevents the progression of LA [122,308,309]. This evidence strongly suggests that intensive BP control interventions are an effective treatment strategy for the management of LA. At the same time, there are some concerns that excessive BP-lowering may lead to harmful effects, including reduced cerebral blood flow and subsequent exacerbated hypoperfusion [310]. However, there is no evidence for this as yet. Most studies have shown that intensive BP control does not decrease cerebral blood flow or affect cerebral perfusion in both hypertensive patients with stroke [311-313] and those without dementia but with extensive CSVD [314], as well as those with dementia [310,315,316]. On the contrary, intensive BP-lowing may increase cerebral blood flow [316,317]. Recently, a SPRINT-MIND substudy found a significant association of intensive BP control to less than 120 mm Hg with increase of cerebral blood flow in white matter and the whole brain in hypertension patients but not with decreased cerebral perfusion, especially in those with a history of cardiovascular disease [318]. Thus, intensive BP-lowering interventions are not expected to have significant effects on cerebral hypoperfusion. Although it seems to be a best intervention for LA as described above, intensive BP-lowing has also been shown to lead to some adverse events in a few studies, such as increased kidney function decline [319], decrease in total brain volume [124,320], and increased risk of dementia [321-324]. Of those detrimental effects, increased dementia risk resulting from the BP-lowing still remains confusing due to inconsistent findings. A few studies showed that the risk of developing dementia increased during the persistent BPlowing in community-dwelling persons over age 75,321 and the steep decline of BP from mid- to late life in older adults (over 65 years) with prehypertension or normotension [322]. It was also observed in the intensive BP treatment in patients with atrial fibrillation or depression [323,324]. On the contrary, more studies showed a significant association of aggressive BP-lowing with the reduced risk of dementia in elderly people [325-330], including the community-dwelling persons aged 60 years or older [331,332], hypertensive patients aged 50 years and older [333], and those with hypertension up to 70 years of age [322]. While some studies do not support those influence of BP control on dementia [325,334-337]. Therefore, further clinical studies are still needed to assess and clarify the potential effects of intensive BP-lowing on adverse health outcomes. Neurologist should also be cautious to adopt this therapy in LA patients, especially in those older individuals.
Taken together, BP control intervention represents an appropriate strategy for the management of LA despite a few harmful effects on kidney function and cognitive function. We believed that intensive BP-lowing therapy could be extensively used in the personalized treatment of LA in the future if those adverse events are controlled well in specific individuals.

Glycemic control

Although the relationship between diabetes mellitus and LA remains controversial as described above, several recent studies have investigated the effect of glycemic control for diabetes mellitus on LA progression [306,338,339]. Of those studies, a double-blind RCT assessing the effects of insulin therapy on white matter health found that intranasal insulin treatment for 12 months significantly reduced the progression of WMH in deep and frontal regions with a similar trend for global LA volume [338]. In addition, one prior study revealed that poor glycemic control was significantly associated with higher WMH burden in patients with APOE4 genotype carriers with type 2 diabetes mellitus, suggesting that the effects of long-term glycemic control on LA in diabetes mellitus were modified by genetic factors [340]. On the contrary, the ACCORD-MIND trial found that the intensive glucose-lowering therapy led to significantly more abnormal white matter volume at 40 months than standard glycemic control, particularly in patients aged less than 60 years [341]. In addition, an observational extension study of ACCORD-MIND (ACCORDION MIND trial) also identified significantly quicker increase in abnormal white matter volume at 40 months in the intensive glucose control group than that in the conventional glucose control group, and observed no significant difference in WMH between the groups at 80 months [302]. Consistently, both a cross-sectional retrospective study and a secondary study of the ACCORD-MIND trial also failed to find the significant association of glycemic control with LA [306,339]. To date, the efficacy of glycemic control on LA remains controversial [342]. There seems to be more evidence suggesting against glucose lowering in patients with SVD [342]. Thus, glycemic control should be cautiously managed in the treatment of LA.

Lipid control

As a vascular risk factor, dyslipidemia (such as hyperlipidemia, higher HDL-C level) is strongly associated with CSVD [118]. Statins are main lipid-lowering drugs wildly used in the prevention and treatment of cardiovascular diseases. They have been shown to be beneficial in the management of CSVD (such as stroke) [343-345]. Thus, statins are also suggested to intervene in the course of LA although the roles of hyperlipidemia and HDL-C in LA remain uncertain to date. Several previous studies investigated the effect of statins on the treatment of LA, but failed to find a beneficial effect of statins upon preventing the progression of LA [96,346-348]. Recently, an 18-month RCT of simvastatin in healthy, statin-naive, cognitively unimpaired, middle-aged adults did also not observe significant effect of simvastatin treatment on WMH lesion volume [349]. However, a substudy of the Cardiovascular Risk Factors and Aging and Incidence of Dementia MRI study found that lipid-lowering drugs decreased the risk of having more severe WMH at late life [105]. Consistently, another randomized, double-blind, placebo-controlled study on the effect of statins on middle cerebral artery stenosis progression among stroke-free individuals also found that lipid-lowering treatment (simvastatin) could delay the progression of cerebral WMH only among those who already have high WMH burden at baseline [350]. Recently, emerging evidence from several studies in Chinese population supports the protective effect of statins on LA [351-353]. They showed that the increase in WMH volume and the risk of WMH progression were significantly lower in the rosuvastatin group than in the placebo group [351-353]. These suggested that statin therapy could ameliorate the progression of LA. Moreover, it was shown that rosuvastatin interacted with telmisartan, an antihypertensive drug on reducing the progression of LA [352]. The precise mechanism that mediates the effectiveness of statins therapy for retarding LA progression remains unclear to date. Statins may help to delay the progression of LA through pleiotropic mechanisms including improving endothelial function and cerebral vasoreactivity [354-356], attenuating inflammatory response [357,358], and decreasing oxidative stress [358,359].
Although more evidence described above seem to suggest statins as an efficient treatment against LA, the lipid-lowering treatment may have to be cautiously managed due to some conflicting findings that statin treatment was associated with increased risk of LA worsening [91,360]. These inconsistent results may be related to blood lipid levels at baseline, baseline WMH, basic disease and pleiotropic effects of statins, even genetic factors. Additionally, statin administration may lead to some adverse effects in the treatment of cerebrovascular disease [360-362], although major protective effects for specific stroke preserve as well [363]. Both the post hoc analysis of the Heart Protection Study and the Stroke Prevention by Aggressive Reduction in Cholesterol Levels trial revealed the association of statin treatment with increased incidence of hemorrhagic stroke [361,362]. Recently, a post hoc analysis on the data from the Japan Statin Treatment Against Recurrent Stroke (J-STARS) study also showed that statins have different influences on the risks of stroke subtypes according to post-randomized LDL-C levels, and they could increase the risk of lacunar stroke in patients [364]. Thus, the use of lipid-lowing drugs in clinical practice should be cautious [342,360]. More studies are still required to assess the efficacy of lipid-modifying therapy strategies on LA in patients with different disease subtypes or disease background.

Homocysteine-lowering therapy

Homocysteine-lowering therapy through multivitamins supplementation is considered as a potential approach for preventing LA. Previously, one study showed that a patient with adult-onset hyperhomocysteinemia due to a vitamin B12 metabolic deficit received homocysteine-lowering treatment, and showed both improved cognitive functions and decreased cortical WMHs at follow-up 18 months [365]. Another study, the VITAmins TO Prevent Stroke (VITATOPS) MRI-Substudy, assessed the effect of vitamins on LA, and found that daily vitamin B administration for two years significantly reduced the progression of LA in those patients with recent stroke or transient ischemic attack and severe CSVD at baseline [366]. Those evidence suggested homocysteine-lowering therapy as an effective treatment method of LA. It could prevent the progression of LA through likely reducing the endothelial dysfunction caused by hyperhomocysteinemia and improving the myelin formation or white matter integrity [190,224,225,367,368].

Antiplatelet therapy

Antiplatelet drugs, such as aspirin, are often used in the prevention and treatment of recurrent stroke [369,370]. As an MRI indicator of CSVD linked to stroke, cognitive decline, and dementia [371], LA may benefit from the antiplatelet agents. Previous study has shown that aspirin, an antiplatelet drug can directly target oligodendroglia cells and promote their differentiation through inhibiting Wnt/β-catenin signaling pathway in vitro and in vivo [372]. Moreover, it has also been shown to promote oligodendrogenesis and oligodendrocyte myelination through extracellular signal-regulated kinase and Ras homolog gene family member A pathways, and improve the learning and memory ability in a well-established WMH model induced by chronic cerebral hypoperfusion [373]. These evidence strongly suggested that aspirin therapy may also represent an effective approach for the treatment of LA with demyelination. However, the clinical studies on the effects of aspirin on the prevention or treatment of LA are rare to date. The only clinical study—Women’s Health Initiative Memory Study of Magnetic Resonance Imaging (WHIMS-MRI) study—found that aspirin use did not have a promisingly positive effect on preventing WMH [374]. Currently, another RCT, the ASPirin in Reducing Events in the Elderly (ASPREE)-NEURO study, is underway to evaluate the effects of low-dose aspirin on LA in the generally healthy elderly [375]. Results of this trial are worth looking forward to being published in the near future. Due to the lack of positive evidence to date, it should be cautious to adopt aspirin therapy in the management of LA.
Compared to aspirin which often causes bleeding complications [376], another common antiplatelet agent-cilostazol-is shown to lead to less hemorrhagic events among patients with ischemic stroke [377,378]. The double-blind RCT, Comparison Study of Cilostazol and Aspirin on Changes in Volume of Cerebral Small Vessel Disease White Matter Changes (CHALLENGE), compared the effects of cilostazol and aspirin on LA progression in patients with CSVD, and found no significant difference in the impact on the progression of LA from baseline to 2 years between two antiplatelet agents [379,380]. It may indicate no efficacy of cilostazol in the prevention or treatment of LA. Recently, another single-center, randomized, double-blind, placebo-controlled study named as DREAM trial (efficacy and safety of cilostazol in DecREasing progression of cerebral WMH) directly investigated the efficacy and safety of cilostazol in preventing CSVD progression but failed to find positive effects of cilostazol treatment on preventing LA progression compared to placebo in stroke- and dementia-free subjects with moderate-to-severe LA [381]. These results do not seem to support the use of cilostazol in the treatment of LA.
Taken together, both cilostazol and aspirin do not seem to have positive effects on slowing down the progression of LA. Given the limited evidence supporting the beneficial effects of antiplatelet agents in LA, we do not recommend using cilostazol and aspirin to manage LA. Future clinical studies will be needed to investigate the efficacy of antiplatelet agents in preventing the progression of LA.

Lifestyle-related interventions

In addition to the drug therapies described above, lifestyle-related interventions such as smoking cessation, maintaining a healthy diet, exercising regularly, avoiding obesity and improving educational levels may also help to prevent LA, manage its potential risk factors, and facilitate treatment.
However, only a few clinical studies support the efficacy of smoking cessation in reducing LA incidence and delaying its progression. An extension of the Atherosclerosis Risk in Communities Study examined the effect of smoking status and history on LA progression, but found no association between WMH progression and either time since smoking cessation or age at smoking initiation [154]. Similarly, another cross-sectional study found no significant relationship between time since smoking cessation and DTI parameters in WMLs, but did report that subjects who had not smoked for more than 20 years had normal-appearing white matter with FA and mean diffusivity values similar to those of subjects who had never smoked [382]. Given the potential significance of smoking cessation in reversing impairments in the structural integrity of the brain, as described previously, we think that the potential benefits of smoking cessation in preventing LA progression need to be explored further.
Dietary habits have been implicated in many human diseases, including CSVDs [383-385]; similarly, a healthy diet is considered to have protective benefits against LA. Two studies—a population-based longitudinal study and a cross-sectional study—showed that dietary interventions can decrease WMH severity and delay LA progression [386,387]. However, a recent two-site RCT involving older adults without cognitive impairment but with a family history of dementia found that change in WMHs from baseline to year 3 did not differ significantly between those who followed the Mediterranean-DASH Intervention for Neurodegenerative Delay diet and those who followed the control diet with mild caloric restriction [388].
As with dietary interventions, physical activity interventions for LA have yielded conflicting results. Five studies, including the recent Australian Imaging Biomarkers and Lifestyle Flagship Study of Aging Active trial [389], showed no significant relationships between physical activity and WMH in older adults [390-393]. However, in four other studies, higher physical activity level was associated with fewer WMHs in individuals without advanced disease, suggesting a beneficial effect of increased physical activity on stopping or slowing the progression of LA [394-397]. Conversely, a longitudinal study found that higher levels of physical activity were associated with LA progression, indicating that it can have detrimental effects as well [398]. It is possible that these conflicting findings indicate that lifestyle interventions represent an indirect strategy for controlling the vascular risk factors for LA. Therefore, interventions related to just one lifestyle factor will not have a significant influence on WMH burden, because LA is influenced by multiple risk factors. Thus, combined interventions targeting multiple lifestyle factors would be more beneficial in LA.
Recently, a cross-sectional analysis of data from the PolyvasculaR Evaluation for Cognitive Impairment and vaScular Events study, which comprehensively controlled for multiple lifestyle factors including diet, physical activity, smoking, alcohol consumption, and BMI [399], showed that participants who adopted four or five low-risk lifestyle habits had lower WMH volumes than those with zero or one low-risk habits. As with the prospective analysis of UK Biobank data, this suggests a significant association between a healthier lifestyle and lower WMH burden in middle-aged and older adults [399]. Thus, comprehensive management of modifiable lifestyle factors should be considered for LA prevention and treatment, although further studies are required to test the efficiency of these interventions in actual patients.

Conclusion and perspectives

Although we have known about the concept of LA for about three decades, its significance in the clinical and academic fields is somewhat underestimated by physicians and neurologists compared to those age-related neurodegenerative conditions such as Parkinson’s disease, Alzheimer’s disease, and stroke. With improvements in imaging technologies and the widespread use of FLAIR-MRI in community hospitals, as well as the increased rate of aging in society, more and more cases of LA are being encountered. This has also led to an increased public awareness about LA. This review article provides an overview of the current advances related to LA for professionals as well as the general public. A better understanding of the imaging heterogeneity, histopathological characteristics, and clinical risk factors of LA will greatly promote our understanding of its nature. The significant associations between certain risk factors (such as age, hypertension, elevated homocysteine level, arterial stiffness) with LA incidence not only offer specific clues regarding its pathogenesis, but also suggest potential intervention strategies for its management. In view of the consistently positive results from clinical association studies and prospective RCTs, controlling key cardiovascular risk factors (such as BP, blood lipid levels) and maintaining a healthy lifestyle (by smoking cessation, physical exercise, healthy diet) should be efficient strategies for preventing the onset and progression of LA.
Additionally, although the heterogeneous histopathological and imaging characteristics of WMHs provide key clues regarding etiology, the underlying molecular mechanisms in LA remain unclear. Genetic studies (especially GWASs) have identified some susceptibility genes for LA and its subclasses in the past decade, which also offer many possibilities for exploring its pathogenesis. Genomic and molecular pathological mechanisms specific to LA subtypes may also represent promising research avenues in the future. Similarly, the identification of biological processes and signaling pathways specific to LA subtypes or its onset and development will help to clarify its pathogenesis and provide potential drug targets and intervention strategies for treatment. This is critical for reducing the public health burden and will benefit aging societies all around the world. Thus, further efforts are needed to construct comprehensive LA models and conduct genetic studies on LA in animals in parallel with clinical studies.

Notes

Funding statement
This study was supported by the Training Program of the National Natural Science Foundation of China in Shanghai Children’s Hospital (Grant No. 2022YGZM05), Young Talent Training Qingmiao Program in Shanghai Children’s Hospital (Grant No. 2022QM2886), and a Fujian Provincial Natural Science Foundation (Grant No. 2022J011365).
Conflicts of interest
The authors have no financial conflicts of interest.
Author contribution
Conceptualization: WQH, QL, CMT. Study design: WQH, CMT. Methodology: WQH, CMT. Data collection: WQH, QL. Investigation: WQH, QL. Statistical analysis: WQH. Writing—original draft: WQH. Writing—review & editing: WQH. Funding acquisition: WQH, QL.

Figure 1.
Global leukoaraiosis epidemiology.
jos-2023-02719f1.jpg
Figure 2.
LA definition and heterogeneous forms on FLAIR-MRI. WMH, white matter hyperintensity; LA, leukoaraiosis; FLAIR-MRI, fluid-attenuated inversionrecovery magnetic resonance imaging.
jos-2023-02719f2.jpg
Figure 3.
Potential risk factors for leukoaraiosis. BP, blood pressure; tHcy, total plasma homocysteine.
jos-2023-02719f3.jpg
Figure 4.
LA susceptibility genes. (A) Risk genes of LA revealed by the previous genetic studies. (B) Venn diagram of risk genes identified by the 7 previous GWAS studies on LA. (C) Venn diagram of risk genes identified by the 18 CGAS, 7 GWAS, and 2 WES studies of LA. (D) Venn diagram of LA subtype-associated risk genes revealed by a genome-wide association meta-analysis of PVWMH, and DWMH. (E) Shared risk genes between PVWMH and DWMH, and its specific susceptibility genes. (F) Venn diagram of dysregulated genes identified by 4 previous gene expression studies of LA [292-295]. (G) Venn diagram of dysregulated and variant genes identified in the blood or lesional tissue of PVWMH, DWMH, and WMH patients, respectively. CGAS, candidate gene association study; GWAS, genome-wide association study; WES, whole-exome sequencing study; DWMHs, deep/subcortical white matter hyperintensity; PVWMH, periventricular white matter hyperintensity; WMH, white matter hyperintensity; LA, leukoaraiosis.
jos-2023-02719f4.jpg

References

1. Hachinski VC, Potter P, Merskey H. Leuko-araiosis. Arch Neurol 1987;44:21-23.
crossref pmid
2. O’Sullivan M. Leukoaraiosis. Pract Neurol 2008;8:26-38.
crossref pmid
3. Hachinski VC, Potter P, Merskey H. Leuko-araiosis: an ancient term for a new problem. Can J Neurol Sci 1986;13(4 Suppl):533-534.
crossref pmid
4. Longstreth WT Jr, Manolio TA, Arnold A, Burke GL, Bryan N, Jungreis CA, et al. Clinical correlates of white matter findings on cranial magnetic resonance imaging of 3301 elderly people. The cardiovascular health study. Stroke 1996;27:1274-1282.
crossref pmid
5. Lin Q, Huang WQ, Ma QL, Lu CX, Tong SJ, Ye JH, et al. Incidence and risk factors of leukoaraiosis from 4683 hospitalized patients: a cross-sectional study. Medicine (Baltimore) 2017;96:e7682.
pmid pmc
6. Wen W, Sachdev PS, Li JJ, Chen X, Anstey KJ. White matter hyperintensities in the forties: their prevalence and topography in an epidemiological sample aged 44-48. Hum Brain Mapp 2009;30:1155-1167.
crossref pmid pmc
7. Ylikoski A, Erkinjuntti T, Raininko R, Sarna S, Sulkava R, Tilvis R. White matter hyperintensities on MRI in the neurologically nondiseased elderly. Analysis of cohorts of consecutive subjects aged 55 to 85 years living at home. Stroke 1995;26:1171-1177.
crossref pmid
8. Dufouil C, Godin O, Chalmers J, Coskun O, MacMahon S, Tzourio-Mazoyer N, et al. Severe cerebral white matter hyperintensities predict severe cognitive decline in patients with cerebrovascular disease history. Stroke 2009;40:2219-2221.
crossref pmid
9. van den Heuvel DM, ten Dam VH, de Craen AJ, AdmiraalBehloul F, Olofsen H, Bollen EL, et al. Increase in periventricular white matter hyperintensities parallels decline in mental processing speed in a non-demented elderly population. J Neurol Neurosurg Psychiatry 2006;77:149-153.
crossref pmid pmc
10. Zheng JJ, Delbaere K, Close JC, Sachdev PS, Lord SR. Impact of white matter lesions on physical functioning and fall risk in older people: a systematic review. Stroke 2011;42:2086-2090.
crossref pmid
11. Moon SY, de Souto Barreto P, Rolland Y, Chupin M, Bouyahia A, Fillon L, et al. Prospective associations between white matter hyperintensities and lower extremity function. Neurology 2018;90:e1291-e1297.
crossref pmid
12. Lee Y, Ko J, Choi YE, Oh JS, Kim JS, Sunwoo MK, et al. Areas of white matter hyperintensities and motor symptoms of Parkinson disease. Neurology 2020;95:e291-e298.
crossref pmid
13. Clancy U, Gilmartin D, Jochems ACC, Knox L, Doubal FN, Wardlaw JM. Neuropsychiatric symptoms associated with cerebral small vessel disease: a systematic review and metaanalysis. Lancet Psychiatry 2021;8:225-236.
crossref pmid
14. Tosto G, Zimmerman ME, Carmichael OT, Brickman AM; Alzheimer’s Disease Neuroimaging Initiative. Predicting aggressive decline in mild cognitive impairment: the importance of white matter hyperintensities. JAMA Neurol 2014;71:872-877.
crossref pmid pmc
15. Kloppenborg RP, Nederkoorn PJ, Geerlings MI, van den Berg E. Presence and progression of white matter hyperintensities and cognition: a meta-analysis. Neurology 2014;82:2127-2138.
crossref pmid
16. Prins ND, Scheltens P. White matter hyperintensities, cognitive impairment and dementia: an update. Nat Rev Neurol 2015;11:157-165.
crossref pmid pdf
17. Hu HY, Ou YN, Shen XN, Qu Y, Ma YH, Wang ZT, et al. White matter hyperintensities and risks of cognitive impairment and dementia: a systematic review and meta-analysis of 36 prospective studies. Neurosci Biobehav Rev 2021;120:16-27.
crossref pmid
18. Whitman GT, Tang Y, Lin A, Baloh RW. A prospective study of cerebral white matter abnormalities in older people with gait dysfunction. Neurology 2001;57:990-994.
crossref pmid
19. Srikanth V, Beare R, Blizzard L, Phan T, Stapleton J, Chen J, et al. Cerebral white matter lesions, gait, and the risk of incident falls: a prospective population-based study. Stroke 2009;40:175-180.
crossref pmid
20. Su C, Yang X, Wei S, Zhao R. Association of cerebral small vessel disease with gait and balance disorders. Front Aging Neurosci 2022;14:834496.
crossref pmid pmc
21. Debette S, Markus HS. The clinical importance of white matter hyperintensities on brain magnetic resonance imaging: systematic review and meta-analysis. BMJ 2010;341:c3666.
crossref pmid pmc
22. Ryu WS, Woo SH, Schellingerhout D, Jang MU, Park KJ, Hong KS, et al. Stroke outcomes are worse with larger leukoaraiosis volumes. Brain 2017;140:158-170.
crossref pmid pmc
23. Georgakis MK, Duering M, Wardlaw JM, Dichgans M. WMH and long-term outcomes in ischemic stroke: a systematic review and meta-analysis. Neurology 2019;92:e1298-e1308.
pmid
24. Imaizumi T, Inamura S, Nomura T. The severities of white matter lesions possibly influence the recurrences of several stroke types. J Stroke Cerebrovasc Dis 2014;23:1897-1902.
crossref pmid
25. Rensma SP, van Sloten TT, Launer LJ, Stehouwer CDA. Cerebral small vessel disease and risk of incident stroke, dementia and depression, and all-cause mortality: a systematic review and meta-analysis. Neurosci Biobehav Rev 2018;90:164-173.
crossref pmid pmc
26. Ghaznawi R, Geerlings MI, Jaarsma-Coes M, Hendrikse J, de Bresser J; UCC-Smart Study Group. Association of white matter hyperintensity markers on MRI and long-term risk of mortality and ischemic stroke: the SMART-MR study. Neurology 2021;96:e2172-e2183.
pmid pmc
27. Godin O, Dufouil C, Maillard P, Delcroix N, Mazoyer B, Crivello F, et al. White matter lesions as a predictor of depression in the elderly: the 3C-Dijon study. Biol Psychiatry 2008;63:663-669.
crossref pmid
28. Herrmann LL, Le Masurier M, Ebmeier KP. White matter hyperintensities in late life depression: a systematic review. J Neurol Neurosurg Psychiatry 2008;79:619-624.
crossref pmid
29. Wang L, Leonards CO, Sterzer P, Ebinger M. White matter lesions and depression: a systematic review and meta-analysis. J Psychiatr Res 2014;56:56-64.
crossref pmid
30. Fang Y, Qin T, Liu W, Ran L, Yang Y, Huang H, et al. Cerebral small-vessel disease and risk of incidence of depression: a meta-analysis of longitudinal cohort studies. J Am Heart Assoc 2020;9:e016512.
crossref pmid pmc
31. Launer LJ, Berger K, Breteler MM, Dufouil C, Fuhrer R, Giampaoli S, et al. Regional variability in the prevalence of cerebral white matter lesions: an MRI study in 9 European countries (CASCADE). Neuroepidemiology 2006;26:23-29.
crossref pmid pdf
32. Mok V, Srikanth V, Xiong Y, Phan TG, Moran C, Chu S, et al. Race-ethnicity and cerebral small vessel disease--comparison between Chinese and White populations. Int J Stroke 2014;9(Suppl A100):36-42.
crossref pmid pdf
33. de Leeuw FE, de Groot JC, Achten E, Oudkerk M, Ramos LM, Heijboer R, et al. Prevalence of cerebral white matter lesions in elderly people: a population based magnetic resonance imaging study. The Rotterdam scan study. J Neurol Neurosurg Psychiatry 2001;70:9-14.
crossref pmid pmc
34. Wen W, Sachdev P. The topography of white matter hyperintensities on brain MRI in healthy 60- to 64-year-old individuals. Neuroimage 2004;22:144-154.
crossref pmid
35. Liao D, Cooper L, Cai J, Toole J, Bryan N, Burke G, et al. The prevalence and severity of white matter lesions, their relationship with age, ethnicity, gender, and cardiovascular disease risk factors: the ARIC study. Neuroepidemiology 1997;16:149-162.
crossref pmid pdf
36. Lam BYK, Yiu B, Ampil E, Chen CL, Dikot Y, Dominguez JC, et al. High burden of cerebral white matter lesion in 9 Asian cities. Sci Rep 2021;11:11587.
crossref pmid pmc pdf
37. Zhang S, Kang X. Investigation of the risk factors for leukoaraiosis (LA). Asia Pac J Public Health 2013;25(4 Suppl):64S71S.
crossref pdf
38. Han F, Zhai FF, Wang Q, Zhou LX, Ni J, Yao M, et al. Prevalence and risk factors of cerebral small vessel disease in a Chinese population-based sample. J Stroke 2018;20:239-246.
crossref pmid pmc pdf
39. Jin H, Ding Z, Lian S, Zhao Y, He S, Zhou L, et al. Prevalence and risk factors of white matter lesions in Tibetan patients without acute stroke. Stroke 2020;51:149-153.
crossref pmid
40. Verny M, Duyckaerts C, Pierot L, Hauw JJ. Leuko-araiosis. Dev Neurosci 1991;13:245-250.
crossref pmid pdf
41. Wahlund LO, Barkhof F, Fazekas F, Bronge L, Augustin M, Sjögren M, et al. A new rating scale for age-related white matter changes applicable to MRI and CT. Stroke 2001;32:1318-1322.
crossref pmid
42. Xiong YY, Mok V. Age-related white matter changes. J Aging Res 2011;2011:617927.
crossref pmid pmc pdf
43. Fazekas F, Barkhof F, Wahlund LO, Pantoni L, Erkinjuntti T, Scheltens P, et al. CT and MRI rating of white matter lesions. Cerebrovasc Dis 2002;13(Suppl 2):31-36.
crossref pdf
44. Kates R, Atkinson D, Brant-Zawadzki M. Fluid-attenuated inversion recovery (FLAIR): clinical prospectus of current and future applications. Top Magn Reson Imaging 1996;8:389-396.
pmid
45. Caligiuri ME, Perrotta P, Augimeri A, Rocca F, Quattrone A, Cherubini A. Automatic detection of white matter hyperintensities in healthy aging and pathology using magnetic resonance imaging: a review. Neuroinformatics 2015;13:261-276.
crossref pmid pmc pdf
46. Tomura N, Kato K, Takahashi S, Sashi R, Sakuma I, Narita K, et al. Comparison of multishot echo-planar fluid-attenuated inversion-recovery imaging with fast spin-echo fluid-attenuated inversion-recovery and T2-weighted imaging in depiction of white matter lesions. J Comput Assist Tomogr 2002;26:810-814.
crossref pmid
47. Piguet O, Ridley LJ, Grayson DA, Bennett HP, Creasey H, Lye TC, et al. Comparing white matter lesions on T2 and FLAIR MRI in the Sydney older persons study. Eur J Neurol 2005;12:399-402.
crossref pmid
48. Barkhof F, Scheltens P. Imaging of white matter lesions. Cerebrovasc Dis 2002;13(Suppl 2):21-30.
crossref pdf
49. Grueter BE, Schulz UG. Age-related cerebral white matter disease (leukoaraiosis): a review. Postgrad Med J 2012;88:79-87.
crossref pmid pdf
50. Taylor WD, Hsu E, Krishnan KR, MacFall JR. Diffusion tensor imaging: background, potential, and utility in psychiatric research. Biol Psychiatry 2004;55:201-207.
crossref pmid
51. Assaf Y, Pasternak O. Diffusion tensor imaging (DTI)-based white matter mapping in brain research: a review. J Mol Neurosci 2008;34:51-61.
crossref pmid pdf
52. Jones DK, Lythgoe D, Horsfield MA, Simmons A, Williams SC, Markus HS. Characterization of white matter damage in ischemic leukoaraiosis with diffusion tensor MRI. Stroke 1999;30:393-397.
crossref pmid
53. Haacke EM, Ayaz M, Khan A, Manova ES, Krishnamurthy B, Gollapalli L, et al. Establishing a baseline phase behavior in magnetic resonance imaging to determine normal vs. abnormal iron content in the brain. J Magn Reson Imaging 2007;26:256-264.
crossref pmid
54. Kraft E, Trenkwalder C, Auer DP. T2*-weighted MRI differentiates multiple system atrophy from Parkinson’s disease. Neurology 2002;59:1265-1267.
crossref pmid
55. Yates PA, Villemagne VL, Ellis KA, Desmond PM, Masters CL, Rowe CC. Cerebral microbleeds: a review of clinical, genetic, and neuroimaging associations. Front Neurol 2014;4:205.
crossref pmid pmc
56. Huang WQ, Lin HN, Lin Q, Tzeng CM. Susceptibility weighted imaging (SWI) recommended as a regular magnetic resonance diagnosis for vascular dementia to identify independent idiopathic normal pressure hydrocephalus before ventriculo-peritoneal (V-P) shunt treatment: a case study. Front Neurol 2019;10:262.
crossref pmid pmc
57. Fazekas F, Chawluk JB, Alavi A, Hurtig HI, Zimmerman RA. MR signal abnormalities at 1.5 T in Alzheimer’s dementia and normal aging. AJR Am J Roentgenol 1987;149:351-356.
crossref pmid
58. Schmidt R, Schmidt H, Haybaeck J, Loitfelder M, Weis S, Cavalieri M, et al. Heterogeneity in age-related white matter changes. Acta Neuropathol 2011;122:171-185.
crossref pmid pdf
59. Kim KW, MacFall JR, Payne ME. Classification of white matter lesions on magnetic resonance imaging in elderly persons. Biol Psychiatry 2008;64:273-280.
crossref pmid pmc
60. Payne ME, Fetzer DL, MacFall JR, Provenzale JM, Byrum CE, Krishnan KR. Development of a semi-automated method for quantification of MRI gray and white matter lesions in geriatric subjects. Psychiatry Res 2002;115:63-77.
crossref pmid
61. Scheltens P, Erkinjunti T, Leys D, Wahlund LO, Inzitari D, del Ser T, et al. White matter changes on CT and MRI: an overview of visual rating scales. Eur Neurol 1998;39:80-89.
crossref pmid pdf
62. Mäntylä R, Erkinjuntti T, Salonen O, Aronen HJ, Peltonen T, Pohjasvaara T, et al. Variable agreement between visual rating scales for white matter hyperintensities on MRI. Comparison of 13 rating scales in a poststroke cohort. Stroke 1997;28:1614-1623.
crossref pmid
63. Kapeller P, Barber R, Vermeulen RJ, Adèr H, Scheltens P, Freidl W, et al. Visual rating of age-related white matter changes on magnetic resonance imaging: scale comparison, interrater agreement, and correlations with quantitative measurements. Stroke 2003;34:441-445.
crossref pmid
64. Admiraal-Behloul F, van den Heuvel DM, Olofsen H, van Osch MJ, van der Grond J, van Buchem MA, et al. Fully automatic segmentation of white matter hyperintensities in MR images of the elderly. Neuroimage 2005;28:607-617.
crossref pmid
65. Scheltens P, Barkhof F, Leys D, Pruvo JP, Nauta JJ, Vermersch P, et al. A semiquantative rating scale for the assessment of signal hyperintensities on magnetic resonance imaging. J Neurol Sci 1993;114:7-12.
crossref pmid
66. Andere A, Jindal G, Molino J, Collins S, Merck D, Burton T, et al. Volumetric white matter hyperintensity ranges correspond to Fazekas scores on brain MRI. J Stroke Cerebrovasc Dis 2022;31:106333.
crossref pmid
67. Lin Q, Huang WQ, Tzeng CM. Genetic associations of leukoaraiosis indicate pathophysiological mechanisms in white matter lesions etiology. Rev Neurosci 2015;26:343-358.
crossref pmid
68. Huang WQ, Yi KH, Li Z, Wang H, Li ML, Cai LL, et al. DNA methylation profiling reveals the change of inflammation-associated ZC3H12D in leukoaraiosis. Front Aging Neurosci 2018;10:143.
crossref pmid pmc
69. Griffanti L, Jenkinson M, Suri S, Zsoldos E, Mahmood A, Filippini N, et al. Classification and characterization of periventricular and deep white matter hyperintensities on MRI: a study in older adults. Neuroimage 2018;170:174-181.
crossref pmid
70. de Groot JC, de Leeuw FE, Oudkerk M, Hofman A, Jolles J, Breteler MM. Cerebral white matter lesions and depressive symptoms in elderly adults. Arch Gen Psychiatry 2000;57:1071-1076.
crossref pmid
71. Krishnan MS, O’Brien JT, Firbank MJ, Pantoni L, Carlucci G, Erkinjuntti T, et al. Relationship between periventricular and deep white matter lesions and depressive symptoms in older people. The LADIS study. Int J Geriatr Psychiatry 2006;21:983-989.
crossref pmid
72. Fazekas F, Kleinert R, Offenbacher H, Schmidt R, Kleinert G, Payer F, et al. Pathologic correlates of incidental MRI white matter signal hyperintensities. Neurology 1993;43:1683-1689.
crossref pmid
73. Prins ND, van Dijk EJ, den Heijer T, Vermeer SE, Koudstaal PJ, Oudkerk M, et al. Cerebral white matter lesions and the risk of dementia. Arch Neurol 2004;61:1531-1534.
crossref pmid
74. de Groot JC, de Leeuw FE, Oudkerk M, van Gijn J, Hofman A, Jolles J, et al. Cerebral white matter lesions and cognitive function: the Rotterdam scan study. Ann Neurol 2000;47:145-151.
crossref pmid
75. De Groot JC, De Leeuw FE, Oudkerk M, Van Gijn J, Hofman A, Jolles J, et al. Periventricular cerebral white matter lesions predict rate of cognitive decline. Ann Neurol 2002;52:335-341.
crossref pmid
76. Sze G, De Armond SJ, Brant-Zawadzki M, Davis RL, Norman D, Newton TH. Foci of MRI signal (pseudo lesions) anterior to the frontal horns: histologic correlations of a normal finding. AJR Am J Roentgenol 1986;147:331-337.
crossref pmid
77. Fazekas F, Schmidt R, Scheltens P. Pathophysiologic mechanisms in the development of age-related white matter changes of the brain. Dement Geriatr Cogn Disord 1998;9(Suppl 1):2-5.
crossref pdf
78. Thomas AJ, O’Brien JT, Barber R, McMeekin W, Perry R. A neuropathological study of periventricular white matter hyperintensities in major depression. J Affect Disord 2003;76:49-54.
crossref pmid
79. van Swieten JC, van den Hout JH, van Ketel BA, Hijdra A, Wokke JH, van Gijn J. Periventricular lesions in the white matter on magnetic resonance imaging in the elderly: a morphometric correlation with arteriolosclerosis and dilated perivascular spaces. Brain 1991;114(Pt 2):761-774.
crossref pmid
80. Wu X, Ya J, Zhou D, Ding Y, Ji X, Meng R. Pathogeneses and imaging features of cerebral white matter lesions of vascular origins. Aging Dis 2021;12:2031-2051.
crossref pmid pmc
81. Mayer PL, Kier EL. The controversy of the periventricular white matter circulation: a review of the anatomic literature. AJNR Am J Neuroradiol 1991;12:223-228.
pmid pmc
82. Moody DM, Bell MA, Challa VR. Features of the cerebral vascular pattern that predict vulnerability to perfusion or oxygenation deficiency: an anatomic study. AJNR Am J Neuroradiol 1990;11:431-439.
pmid pmc
83. Beyer JL, Young R, Kuchibhatla M, Krishnan KR. Hyperintense MRI lesions in bipolar disorder: a meta-analysis and review. Int Rev Psychiatry 2009;21:394-409.
crossref pmid pmc
84. ten Dam VH, van den Heuvel DM, de Craen AJ, Bollen EL, Murray HM, Westendorp RG, et al. Decline in total cerebral blood flow is linked with increase in periventricular but not deep white matter hyperintensities. Radiology 2007;243:198-203.
crossref pmid
85. Stewart PA, Magliocco M, Hayakawa K, Farrell CL, Del Maestro RF, Girvin J, et al. A quantitative analysis of blood-brain barrier ultrastructure in the aging human. Microvasc Res 1987;33:270-282.
crossref pmid
86. Pantoni L, Garcia JH. Pathogenesis of leukoaraiosis: a review. Stroke 1997;28:652-659.
crossref pmid
87. Pantoni L. Pathophysiology of age-related cerebral white matter changes. Cerebrovasc Dis 2002;13(Suppl 2):7-10.
crossref pdf
88. Jung KH, Stephens KA, Yochim KM, Riphagen JM, Kim CM, Buckner RL, et al. Heterogeneity of cerebral white matter lesions and clinical correlates in older adults. Stroke 2021;52:620-630.
crossref pmid pmc
89. Schmidt R, Fazekas F, Kapeller P, Schmidt H, Hartung HP. MRI white matter hyperintensities: three-year follow-up of the Austrian stroke prevention study. Neurology 1999;53:132-139.
crossref pmid
90. Schmidt R, Enzinger C, Ropele S, Schmidt H, Fazekas F; Austrian Stroke Prevention Study. Progression of cerebral white matter lesions: 6-year results of the Austrian stroke prevention study. Lancet 2003;361:2046-2048.
crossref pmid
91. Longstreth WT Jr, Arnold AM, Beauchamp NJ Jr, Manolio TA, Lefkowitz D, Jungreis C, et al. Incidence, manifestations, and predictors of worsening white matter on serial cranial magnetic resonance imaging in the elderly: the cardiovascular health study. Stroke 2005;36:56-61.
crossref pmid
92. Taylor WD, MacFall JR, Provenzale JM, Payne ME, McQuoid DR, Steffens DC, et al. Serial MR imaging of volumes of hyperintense white matter lesions in elderly patients: correlation with vascular risk factors. AJR Am J Roentgenol 2003;181:571-576.
crossref pmid
93. Masana Y, Motozaki T. Emergence and progress of white matter lesion in brain check-up. Acta Neurol Scand 2003;107:187-194.
crossref pmid pdf
94. van Dijk EJ, Prins ND, Vrooman HA, Hofman A, Koudstaal PJ, Breteler MM. Progression of cerebral small vessel disease in relation to risk factors and cognitive consequences: Rotterdam scan study. Stroke 2008;39:2712-2719.
crossref pmid
95. Sachdev P, Wen W, Chen X, Brodaty H. Progression of white matter hyperintensities in elderly individuals over 3 years. Neurology 2007;68:214-222.
crossref pmid
96. Gyanwali B, Shaik MA, Tan BY, Venketasubramanian N, Chen C, Hilal S. Risk factors for and clinical relevance of incident and progression of cerebral small vessel disease markers in an Asian memory clinic population. J Alzheimers Dis 2019;67:1209-1219.
crossref pmid
97. Sachdev P, Chen X, Wen W. White matter hyperintensities in mid-adult life. Curr Opin Psychiatry 2008;21:268-274.
crossref pmid
98. van den Heuvel DM, Admiraal-Behloul F, ten Dam VH, Olofsen H, Bollen EL, Murray HM, et al. Different progression rates for deep white matter hyperintensities in elderly men and women. Neurology 2004;63:1699-1701.
crossref pmid
99. Liao D, Cooper L, Cai J, Toole JF, Bryan NR, Hutchinson RG, et al. Presence and severity of cerebral white matter lesions and hypertension, its treatment, and its control: the ARIC study. Stroke 1996;27:2262-2270.
crossref pmid
100. de Leeuw FE, de Groot JC, Oudkerk M, Witteman JC, Hofman A, van Gijn J, et al. A follow-up study of blood pressure and cerebral white matter lesions. Ann Neurol 1999;46:827-833.
crossref pmid
101. de Leeuw FE, de Groot JC, Oudkerk M, Witteman JC, Hofman A, van Gijn J, et al. Hypertension and cerebral white matter lesions in a prospective cohort study. Brain 2002;125(Pt 4):765-772.
crossref pmid
102. van Dijk EJ, Breteler MM, Schmidt R, Berger K, Nilsson LG, Oudkerk M, et al. The association between blood pressure, hypertension, and cerebral white matter lesions: cardiovascular determinants of dementia study. Hypertension 2004;44:625-630.
crossref pmid
103. Zhao Y, Ke Z, He W, Cai Z. Volume of white matter hyperintensities increases with blood pressure in patients with hypertension. J Int Med Res 2019;47:3681-3689.
crossref pmid pmc pdf
104. Basile AM, Pantoni L, Pracucci G, Asplund K, Chabriat H, Erkinjuntti T, et al. Age, hypertension, and lacunar stroke are the major determinants of the severity of age-related white matter changes: the LADIS (leukoaraiosis and disability in the elderly) study. Cerebrovasc Dis 2006;21:315-322.
crossref pmid pdf
105. Vuorinen M, Solomon A, Rovio S, Nieminen L, Kåreholt I, Tuomilehto J, et al. Changes in vascular risk factors from midlife to late life and white matter lesions: a 20-year follow-up study. Dement Geriatr Cogn Disord 2011;31:119-125.
crossref pmid pdf
106. Habes M, Erus G, Toledo JB, Zhang T, Bryan N, Launer LJ, et al. White matter hyperintensities and imaging patterns of brain ageing in the general population. Brain 2016;139(Pt 4):1164-1179.
crossref pmid pmc
107. Guo X, Pantoni L, Simoni M, Bengtsson C, Björkelund C, Lissner L, et al. Blood pressure components and changes in relation to white matter lesions: a 32-year prospective population study. Hypertension 2009;54:57-62.
crossref pmid
108. Lane CA, Barnes J, Nicholas JM, Sudre CH, Cash DM, Parker TD, et al. Associations between blood pressure across adulthood and late-life brain structure and pathology in the neuroscience substudy of the 1946 British birth cohort (Insight 46): an epidemiological study. Lancet Neurol 2019;18:942-952.
crossref pmid pmc
109. Wartolowska KA, Webb AJS. Midlife blood pressure is associated with the severity of white matter hyperintensities: analysis of the UK Biobank cohort study. Eur Heart J 2021;42:750-757.
crossref pmid pmc pdf
110. Sargurupremraj M, Suzuki H, Jian X, Sarnowski C, Evans TE, Bis JC, et al. Cerebral small vessel disease genomics and its implications across the lifespan. Nat Commun 2020;11:6285.
pmid pmc
111. Wilkinson I, Webb AJS. Consistency of associations of systolic and diastolic blood pressure with white matter hyperintensities: a meta-analysis. Int J Stroke 2022;17:291-298.
crossref pmid pmc pdf
112. Marcus J, Gardener H, Rundek T, Elkind MS, Sacco RL, Decarli C, et al. Baseline and longitudinal increases in diastolic blood pressure are associated with greater white matter hyperintensity volume: the northern Manhattan study. Stroke 2011;42:2639-2641.
crossref pmid pmc
113. Shokouhi M, Qiu D, Samman Tahhan A, Quyyumi AA, Hajjar I. Differential associations of diastolic and systolic pressures with cerebral measures in older individuals with mild cognitive impairment. Am J Hypertens 2018;31:1268-1277.
crossref pmid pmc pdf
114. Sudre CH, Smith L, Atkinson D, Chaturvedi N, Ourselin S, Barkhof F, et al. Cardiovascular risk factors and white matter hyperintensities: difference in susceptibility in South Asians compared with Europeans. J Am Heart Assoc 2018;7:e010533.
crossref pmid pmc
115. Caunca MR, Simonetto M, Cheung YK, Alperin N, Lee SH, Elkind MSV, et al. Diastolic blood pressure is associated with regional white matter lesion load: the northern Manhattan study. Stroke 2020;51:372-378.
crossref pmid pmc
116. Verhaaren BF, Vernooij MW, de Boer R, Hofman A, Niessen WJ, van der Lugt A, et al. High blood pressure and cerebral white matter lesion progression in the general population. Hypertension 2013;61:1354-1359.
crossref pmid
117. Dufouil C, de Kersaint-Gilly A, Besançon V, Levy C, Auffray E, Brunnereau L, et al. Longitudinal study of blood pressure and white matter hyperintensities: the EVA MRI cohort. Neurology 2001;56:921-926.
crossref pmid
118. Pantoni L. Cerebral small vessel disease: from pathogenesis and clinical characteristics to therapeutic challenges. Lancet Neurol 2010;9:689-701.
crossref pmid
119. Sierra C. Essential hypertension, cerebral white matter pathology and ischemic stroke. Curr Med Chem 2014;21:2156-2164.
crossref pmid
120. Dufouil C, Chalmers J, Coskun O, Besançon V, Bousser MG, Guillon P, et al. Effects of blood pressure lowering on cerebral white matter hyperintensities in patients with stroke: the PROGRESS (perindopril protection against recurrent stroke study) magnetic resonance imaging substudy. Circulation 2005;112:1644-1650.
crossref pmid
121. Firbank MJ, Wiseman RM, Burton EJ, Saxby BK, O’Brien JT, Ford GA. Brain atrophy and white matter hyperintensity change in older adults and relationship to blood pressure. Brain atrophy, WMH change and blood pressure. J Neurol 2007;254:713-721.
crossref pmid pdf
122. van Middelaar T, Argillander TE, Schreuder FHBM, Deinum J, Richard E, Klijn CJM. Effect of antihypertensive medication on cerebral small vessel disease: a systematic review and metaanalysis. Stroke 2018;49:1531-1533.
crossref pmid
123. Kjeldsen SE, Narkiewicz K, Burnier M, Oparil S. Intensive blood pressure lowering prevents mild cognitive impairment and possible dementia and slows development of white matter lesions in brain: the SPRINT memory and cognition in decreased hypertension (SPRINT MIND) study. Blood Press 2018;27:247-248.
crossref pmid
124. SPRINT MIND Investigators for the SPRINT Research Group. Association of intensive vs standard blood pressure control with cerebral white matter lesions. JAMA 2019;322:524-534.
pmid pmc
125. Cox SR, Lyall DM, Ritchie SJ, Bastin ME, Harris MA, Buchanan CR, et al. Associations between vascular risk factors and brain MRI indices in UK Biobank. Eur Heart J 2019;40:2290-2300.
crossref pmid pmc pdf
126. Veldink JH, Scheltens P, Jonker C, Launer LJ. Progression of cerebral white matter hyperintensities on MRI is related to diastolic blood pressure. Neurology 1998;51:319-320.
crossref pmid
127. Zhang D, Tang Y, Ge J, Liu Y, Jin J, He M. Age and diastolic blood pressure play an important role in the progression of white matter lesions: a meta-analysis. Eur Neurol 2020;83:351-359.
crossref pmid pdf
128. Pantoni L, Garcia JH. The significance of cerebral white matter abnormalities 100 years after Binswanger’s report. A review. Stroke 1995;26:1293-1301.
crossref pmid
129. Topakian R, Barrick TR, Howe FA, Markus HS. Blood-brain barrier permeability is increased in normal-appearing white matter in patients with lacunar stroke and leucoaraiosis. J Neurol Neurosurg Psychiatry 2010;81:192-197.
crossref pmid
130. Oishi E, Ohara T, Sakata S, Fukuhara M, Hata J, Yoshida D, et al. Day-to-day blood pressure variability and risk of dementia in a general Japanese elderly population: the Hisayama study. Circulation 2017;136:516-525.
crossref pmid pmc
131. Stevens SL, Wood S, Koshiaris C, Law K, Glasziou P, Stevens RJ, et al. Blood pressure variability and cardiovascular disease: systematic review and meta-analysis. BMJ 2016;354:i4098.
crossref pmid pmc
132. Alpérovitch A, Blachier M, Soumaré A, Ritchie K, Dartigues JF, Richard-Harston S, et al. Blood pressure variability and risk of dementia in an elderly cohort, the three-city study. Alzheimers Dement 2014;10(5 Suppl):S330-S337.
pmid
133. Gunstad J, Cohen RA, Tate DF, Paul RH, Poppas A, Hoth K, et al. Blood pressure variability and white matter hyperintensities in older adults with cardiovascular disease. Blood Press 2005;14:353-358.
crossref pmid pmc
134. Brickman AM, Reitz C, Luchsinger JA, Manly JJ, Schupf N, Muraskin J, et al. Long-term blood pressure fluctuation and cerebrovascular disease in an elderly cohort. Arch Neurol 2010;67:564-569.
crossref pmid pmc
135. Liu Z, Zhao Y, Zhang H, Chai Q, Cui Y, Diao Y, et al. Excessive variability in systolic blood pressure that is self-measured at home exacerbates the progression of brain white matter lesions and cognitive impairment in the oldest old. Hypertens Res 2016;39:245-253.
crossref pmid pdf
136. Filomena J, Riba-Llena I, Vinyoles E, Tovar JL, Mundet X, Castañé X, et al. Short-term blood pressure variability relates to the presence of subclinical brain small vessel disease in primary hypertension. Hypertension 2015;66:634-640. discussion 445.
crossref pmid
137. Yang S, Yuan J, Qin W, Yang L, Fan H, Li Y, et al. Twenty-four-hour ambulatory blood pressure variability is associated with total magnetic resonance imaging burden of cerebral small-vessel disease. Clin Interv Aging 2018;13:1419-1427.
pmid pmc
138. Chen X, Zhu Y, Geng S, Li Q, Jiang H. Association of blood pressure variability and intima-media thickness with white matter hyperintensities in hypertensive patients. Front Aging Neurosci 2019;11:192.
crossref pmid pmc
139. van Middelaar T, Richard E, Moll van Charante EP, van Gool WA, van Dalen JW. Visit-to-visit blood pressure variability and progression of white matter hyperintensities among older people with hypertension. J Am Med Dir Assoc 2019;20:1175-1177.e1.
crossref pmid
140. Shen J, Yang L, Xu Z, Wei W. Association between twenty-four-hour ambulatory blood pressure variability and cerebral small vessel disease burden in acute ischemic stroke. Behav Neurol 2022;2022:3769577.
crossref pmid pmc pdf
141. Zhang B, Huo Y, Yang Z, Lv H, Wang Y, Feng J, et al. Day to day blood pressure variability associated with cerebral arterial dilation and white matter hyperintensity. Hypertension 2022;79:1455-1465.
crossref pmid pmc
142. Tully PJ, Yano Y, Launer LJ, Kario K, Nagai M, Mooijaart SP, et al. Association between blood pressure variability and cerebral small-vessel disease: a systematic review and meta-analysis. J Am Heart Assoc 2020;9:e013841.
pmid
143. Havlik RJ, Foley DJ, Sayer B, Masaki K, White L, Launer LJ. Variability in midlife systolic blood pressure is related to latelife brain white matter lesions: the Honolulu-Asia aging study. Stroke 2002;33:26-30.
crossref pmid
144. Zhou TL, Rensma SP, van der Heide FCT, Henry RMA, Kroon AA, Houben AJHM, et al. Blood pressure variability and microvascular dysfunction: the Maastricht study. J Hypertens 2020;38:1541-1550.
crossref pmid
145. Jiang X, Guo Y, Zhao Y, Gao X, Peng D, Zhang H, et al. Multiscale dynamics of blood pressure fluctuation is associated with white matter lesion burden in older adults with and without hypertension: observations from a pilot study. Front Cardiovasc Med 2021;8:636702.
crossref pmid pmc
146. Starmans NL, Wolters FJ, Leeuwis AE, Bron EE, Brunner La Rocca HP, Staals J, et al. Twenty-four hour blood pressure variability and the prevalence and the progression of cerebral white matter hyperintensities. J Cereb Blood Flow Metab 2023;43:801-811.
crossref pmid pmc pdf
147. Ma Y, Song A, Viswanathan A, Blacker D, Vernooij MW, Hofman A, et al. Blood pressure variability and cerebral small vessel disease: a systematic review and meta-analysis of population-based cohorts. Stroke 2020;51:82-89.
crossref pmid pmc
148. Jeerakathil T, Wolf PA, Beiser A, Massaro J, Seshadri S, D’Agostino RB, et al. Stroke risk profile predicts white matter hyperintensity volume: the Framingham study. Stroke 2004;35:1857-1861.
crossref pmid
149. Jongen C, van der Grond J, Kappelle LJ, Biessels GJ, Viergever MA, Pluim JP; Utrecht Diabetic Encephalopathy Study Group. Automated measurement of brain and white matter lesion volume in type 2 diabetes mellitus. Diabetologia 2007;50:1509-1516.
crossref pmid pmc pdf
150. King KS, Vintimilla RM, Braskie MN, Wei K, Hall JR, Borzage M, et al. Vascular risk profile and white matter hyperintensity volume among Mexican Americans and non-Hispanic Whites: the HABLE study. Alzheimers Dement (Amst) 2022;14:e12263.
crossref pmid pmc pdf
151. Guan J, Yan C, Gao Q, Li J, Wang L, Hong M, et al. Analysis of risk factors in patients with leukoaraiosis. Medicine (Baltimore) 2017;96:e6153.
crossref pmid pmc
152. Hilal S, Mok V, Youn YC, Wong A, Ikram MK, Chen CL. Prevalence, risk factors and consequences of cerebral small vessel diseases: data from three Asian countries. J Neurol Neurosurg Psychiatry 2017;88:669-674.
crossref pmid
153. Debette S, Seshadri S, Beiser A, Au R, Himali JJ, Palumbo C, et al. Midlife vascular risk factor exposure accelerates structural brain aging and cognitive decline. Neurology 2011;77:461-468.
crossref pmid pmc
154. Power MC, Deal JA, Sharrett AR, Jack CR Jr, Knopman D, Mosley TH, et al. Smoking and white matter hyperintensity progression: the ARIC-MRI study. Neurology 2015;84:841-848.
crossref pmid pmc
155. Das AS, Regenhardt RW, Vernooij MW, Blacker D, Charidimou A, Viswanathan A. Asymptomatic cerebral small vessel disease: insights from population-based studies. J Stroke 2019;21:121-138.
crossref pmid pmc pdf
156. Jimenez-Conde J, Biffi A, Rahman R, Kanakis A, Butler C, Sonni S, et al. Hyperlipidemia and reduced white matter hyperintensity volume in patients with ischemic stroke. Stroke 2010;41:437-442.
crossref pmid pmc
157. Ohwaki K, Yano E, Tamura A, Inoue T, Saito I. Hypercholesterolemia is associated with a lower risk of cerebral ischemic small vessel disease detected on brain checkups. Clin Neurol Neurosurg 2013;115:669-672.
crossref pmid
158. Ke D, Zhou F, Liang H, Xu Y, Lou H. Hypertriglyceridemia is associated with reduced leukoaraiosis severity in patients with a small vessel stroke. Behav Neurol 2018;2018:1361780.
crossref pmid pmc pdf
159. Wang Z, Chen Q, Chen J, Yang N, Zheng K. Risk factors of cerebral small vessel disease: a systematic review and metaanalysis. Medicine (Baltimore) 2021;100:e28229.
pmid pmc
160. Okamura T, Hashimoto Y, Hamaguchi M, Ohbora A, Kojima T, Fukui M. Metabolically healthy obesity and risk of leukoaraiosis; a population based cross-sectional study. Endocr J 2018;65:669-675.
crossref pmid
161. Coutinho T, Turner ST, Kullo IJ. Aortic pulse wave velocity is associated with measures of subclinical target organ damage. JACC Cardiovasc Imaging 2011;4:754-761.
crossref pmid pmc
162. Mitchell GF, van Buchem MA, Sigurdsson S, Gotal JD, Jonsdottir MK, Kjartansson Ó, et al. Arterial stiffness, pressure and flow pulsatility and brain structure and function: the age, gene/environment susceptibility--Reykjavik study. Brain 2011;134(Pt 11):3398-3407.
crossref pmid pmc
163. Poels MM, Zaccai K, Verwoert GC, Vernooij MW, Hofman A, van der Lugt A, et al. Arterial stiffness and cerebral small vessel disease: the Rotterdam scan study. Stroke 2012;43:2637-2642.
crossref pmid
164. Saji N, Shimizu H, Kawarai T, Tadano M, Kita Y, Yokono K. Increased brachial-ankle pulse wave velocity is independently associated with white matter hyperintensities. Neuroepidemiology 2011;36:252-257.
crossref pmid pdf
165. Tsao CW, Seshadri S, Beiser AS, Westwood AJ, Decarli C, Au R, et al. Relations of arterial stiffness and endothelial function to brain aging in the community. Neurology 2013;81:984-991.
crossref pmid pmc
166. Singer J, Trollor JN, Baune BT, Sachdev PS, Smith E. Arterial stiffness, the brain and cognition: a systematic review. Ageing Res Rev 2014;15:16-27.
crossref pmid
167. van Sloten TT, Protogerou AD, Henry RM, Schram MT, Launer LJ, Stehouwer CD. Association between arterial stiffness, cerebral small vessel disease and cognitive impairment: a systematic review and meta-analysis. Neurosci Biobehav Rev 2015;53:121-130.
crossref pmid pmc
168. Haidegger M, Lindenbeck S, Hofer E, Rodler C, Zweiker R, Perl S, et al. Arterial stiffness and its influence on cerebral morphology and cognitive function. Ther Adv Neurol Disord 2023;16:17562864231180715.
crossref pmid pmc pdf
169. Caughey MC, Qiao Y, Meyer ML, Palta P, Matsushita K, Tanaka H, et al. Relationship between central artery stiffness, brain arterial dilation, and white matter hyperintensities in older adults: the ARIC study—brief report. Arterioscler Thromb Vasc Biol 2021;41:2109-2116.
crossref pmid pmc
170. Robert C, Ling LH, Tan ESJ, Gyanwali B, Venketasubramanian N, Lim SL, et al. Effects of carotid artery stiffness on cerebral small-vessel disease and cognition. J Am Heart Assoc 2022;11:e027295.
crossref pmid pmc
171. Miyagi T, Ishida A, Shinzato T, Ohya Y. Arterial stiffness is associated with small vessel disease irrespective of blood pressure in stroke-free individuals. Stroke 2023;54:2814-2821.
crossref pmid
172. Tomoto T, Tarumi T, Zhang R. Central arterial stiffness, brain white matter hyperintensity and total brain volume across the adult lifespan. J Hypertens 2023;41:819-829.
crossref pmid pmc
173. Brandts A, van Elderen SG, Westenberg JJ, van der Grond J, van Buchem MA, Huisman MV, et al. Association of aortic arch pulse wave velocity with left ventricular mass and lacunar brain infarcts in hypertensive patients: assessment with MR imaging. Radiology 2009;253:681-688.
crossref pmid
174. van Elderen SG, Brandts A, Westenberg JJ, van der Grond J, Tamsma JT, van Buchem MA, et al. Aortic stiffness is associated with cardiac function and cerebral small vessel disease in patients with type 1 diabetes mellitus: assessment by magnetic resonance imaging. Eur Radiol 2010;20:1132-1138.
crossref pmid pmc
175. Ohmine T, Miwa Y, Yao H, Yuzuriha T, Takashima Y, Uchino A, et al. Association between arterial stiffness and cerebral white matter lesions in community-dwelling elderly subjects. Hypertens Res 2008;31:75-81.
crossref pmid
176. Hannawi Y, Vaidya D, Yanek LR, Johansen MC, Kral BG, Becker LC, et al. Association of vascular properties with the brain white matter hyperintensity in middle-aged population. J Am Heart Assoc 2022;11:e024606.
crossref pmid pmc
177. Funck KL, Laugesen E, Høyem P, Stausbøl-Grøn B, Kim WY, Østergaard L, et al. Arterial stiffness and progression of cerebral white matter hyperintensities in patients with type 2 diabetes and matched controls: a 5-year cohort study. Diabetol Metab Syndr 2021;13:71.
crossref pmid pmc pdf
178. Del Brutto OH, Mera RM, Costa AF, Recalde BY, Rumbea DA, Sedler MJ. Arterial stiffness and progression of white matter hyperintensities of presumed vascular origin in communitydwelling older adults of Amerindian ancestry: the Atahualpa project cohort. Clin Neurol Neurosurg 2022;221:107411.
crossref pmid
179. Allison EY, Al-Khazraji BK. Association of arterial stiffness index and brain structure in the UK Biobank: a 10-year retrospective analysis. Aging Dis 2023 Jun 8 [Epub]. https://doi.org/10.14336/AD.2023.0419.
crossref
180. Scheuermann BC, Parr SK, Schulze KM, Kunkel ON, Turpin VG, Liang J, et al. Associations of cerebrovascular regulation and arterial stiffness with cerebral small vessel disease: a systematic review and meta-analysis. J Am Heart Assoc 2023;12:e032616.
crossref pmid pmc
181. Hassan A, Hunt BJ, O’Sullivan M, Bell R, D’Souza R, Jeffery S, et al. Homocysteine is a risk factor for cerebral small vessel disease, acting via endothelial dysfunction. Brain 2004;127(Pt 1):212-219.
crossref pmid
182. Welch GN, Loscalzo J. Homocysteine and atherothrombosis. N Engl J Med 1998;338:1042-1050.
crossref pmid
183. Kamath AF, Chauhan AK, Kisucka J, Dole VS, Loscalzo J, Handy DE, et al. Elevated levels of homocysteine compromise blood-brain barrier integrity in mice. Blood 2006;107:591-593.
crossref pmid pmc pdf
184. Wald DS, Law M, Morris JK. Homocysteine and cardiovascular disease: evidence on causality from a meta-analysis. BMJ 2002;325:1202.
crossref pmid pmc
185. Homocysteine Studies Collaboration. Homocysteine and risk of ischemic heart disease and stroke: a meta-analysis. JAMA 2002;288:2015-2022.
crossref pmid
186. Boushey CJ, Beresford SA, Omenn GS, Motulsky AG. A quantitative assessment of plasma homocysteine as a risk factor for vascular disease. Probable benefits of increasing folic acid intakes. JAMA 1995;274:1049-1057.
crossref pmid
187. Dufouil C, Alpérovitch A, Ducros V, Tzourio C. Homocysteine, white matter hyperintensities, and cognition in healthy elderly people. Ann Neurol 2003;53:214-221.
crossref pmid pdf
188. Longstreth WT Jr, Katz R, Olson J, Bernick C, Carr JJ, Malinow MR, et al. Plasma total homocysteine levels and cranial magnetic resonance imaging findings in elderly persons: the cardiovascular health study. Arch Neurol 2004;61:67-72.
crossref pmid
189. Seshadri S, Wolf PA, Beiser AS, Selhub J, Au R, Jacques PF, et al. Association of plasma total homocysteine levels with subclinical brain injury: cerebral volumes, white matter hyperintensity, and silent brain infarcts at volumetric magnetic resonance imaging in the Framingham Offspring Study. Arch Neurol 2008;65:642-649.
pmid pmc
190. Hogervorst E, Ribeiro HM, Molyneux A, Budge M, Smith AD. Plasma homocysteine levels, cerebrovascular risk factors, and cerebral white matter changes (leukoaraiosis) in patients with Alzheimer disease. Arch Neurol 2002;59:787-793.
crossref pmid
191. Vermeer SE, van Dijk EJ, Koudstaal PJ, Oudkerk M, Hofman A, Clarke R, et al. Homocysteine, silent brain infarcts, and white matter lesions: the Rotterdam scan study. Ann Neurol 2002;51:285-289.
crossref pmid
192. Wright CB, Paik MC, Brown TR, Stabler SP, Allen RH, Sacco RL, et al. Total homocysteine is associated with white matter hyperintensity volume: the northern Manhattan study. Stroke 2005;36:1207-1211.
crossref pmid pmc
193. Naka H, Nomura E, Takahashi T, Wakabayashi S, Kajikawa H, Kohriyama T, et al. Plasma total homocysteine levels are associated with advanced leukoaraiosis but not with asymptomatic microbleeds on T2*-weighted MRI in patients with stroke. Eur J Neurol 2006;13:261-265.
crossref pmid
194. Shimomura T, Anan F, Umeno Y, Eshima N, Saikawa T, Yoshimatsu H, et al. Hyperhomocysteinaemia is a significant risk factor for white matter lesions in Japanese type 2 diabetic patients. Eur J Neurol 2008;15:289-294.
crossref pmid
195. Anan F, Masaki T, Tatsukawa H, Nagano S, Oribe M, Eshima N, et al. The role of homocysteine as a significant risk factor for white matter lesions in Japanese women with rheumatoid arthritis. Metabolism 2009;58:69-73.
crossref pmid
196. Tseng YL, Chang YY, Liu JS, Su CS, Lai SL, Lan MY. Association of plasma homocysteine concentration with cerebral white matter hyperintensity on magnetic resonance images in stroke patients. J Neurol Sci 2009;284:36-39.
crossref pmid
197. Pavlovic AM, Pekmezovic T, Obrenovic R, Novakovic I, Tomic G, Mijajlovic M, et al. Increased total homocysteine level is associated with clinical status and severity of white matter changes in symptomatic patients with subcortical small vessel disease. Clin Neurol Neurosurg 2011;113:711-715.
crossref pmid
198. Raz N, Yang Y, Dahle CL, Land S. Volume of white matter hyperintensities in healthy adults: contribution of age, vascular risk factors, and inflammation-related genetic variants. Biochim Biophys Acta 2012;1822:361-369.
crossref pmid pmc
199. Cloonan L, Fitzpatrick KM, Kanakis AS, Furie KL, Rosand J, Rost NS. Metabolic determinants of white matter hyperintensity burden in patients with ischemic stroke. Atherosclerosis 2015;240:149-153.
crossref pmid pmc
200. Shan Y, Tan S, Wang Y, Li K, Zhang L, Liao S, et al. Risk factors and clinical manifestations of juxtacortical small lesions: a neuroimaging study. Front Neurol 2017;8:497.
crossref pmid pmc
201. Piao X, Wu G, Yang P, Shen J, De A, Wu J, et al. Association between homocysteine and cerebral small vessel disease: a meta-analysis. J Stroke Cerebrovasc Dis 2018;27:2423-2430.
crossref pmid
202. Shen Y, Dong ZF, Pan PL, Xu G, Huang JY, Liu CF. Association of homocysteine, folate, and white matter hyperintensities in Parkinson’s patients with different motor phenotypes. Neurol Sci 2019;40:1855-1863.
crossref pmid pdf
203. Nam KW, Kwon HM, Jeong HY, Park JH, Kwon H, Jeong SM. Serum homocysteine level is related to cerebral small vessel disease in a healthy population. Neurology 2019;92:e317-e325.
crossref pmid
204. Wang X, Yin H, Ji X, Sang S, Shao S, Wang G, et al. Association between homocysteine and white matter hyperintensities in rural-dwelling Chinese people with asymptomatic intracranial arterial stenosis: a population-based study. Brain Behav 2021;11:e02205.
pmid pmc
205. Kloppenborg RP, Geerlings MI, Visseren FL, Mali WP, Vermeulen M, van der Graaf Y, et al. Homocysteine and progression of generalized small-vessel disease: the SMART-MR study. Neurology 2014;82:777-783.
crossref pmid
206. Hooshmand B, Mangialasche F, Kalpouzos G, Solomon A, Kåreholt I, Smith AD, et al. Association of vitamin B12, folate, and sulfur amino acids with brain magnetic resonance imaging measures in older adults: a longitudinal population-based study. JAMA Psychiatry 2016;73:606-613.
crossref pmid
207. Sachdev P, Parslow R, Salonikas C, Lux O, Wen W, Kumar R, et al. Homocysteine and the brain in midadult life: evidence for an increased risk of leukoaraiosis in men. Arch Neurol 2004;61:1369-1376.
crossref pmid
208. Gao Y, Wei S, Song B, Qin J, Fang H, Ji Y, et al. Homocysteine level is associated with white matter hyperintensity locations in patients with acute ischemic stroke. PLoS One 2015;10:e0144431.
crossref pmid pmc
209. Lee KO, Woo MH, Chung D, Choi JW, Kim NK, Kim OJ, et al. Differential impact of plasma homocysteine levels on the periventricular and subcortical white matter hyperintensities on the brain. Front Neurol 2019;10:1174.
crossref pmid pmc
210. Refsum H, Smith AD, Ueland PM, Nexo E, Clarke R, McPartlin J, et al. Facts and recommendations about total homocysteine determinations: an expert opinion. Clin Chem 2004;50:3-32.
crossref pmid pdf
211. Selhub J, Jacques PF, Wilson PW, Rush D, Rosenberg IH. Vitamin status and intake as primary determinants of homocysteinemia in an elderly population. JAMA 1993;270:2693-2698.
crossref pmid
212. Clarke R, Smith AD, Jobst KA, Refsum H, Sutton L, Ueland PM. Folate, vitamin B12, and serum total homocysteine levels in confirmed Alzheimer disease. Arch Neurol 1998;55:1449-1455.
crossref pmid
213. Stabler SP. Vitamin B12 deficiency. N Engl J Med 2013;368:149-160.
crossref pmid
214. Selhub J, Bagley LC, Miller J, Rosenberg IH. B vitamins, homocysteine, and neurocognitive function in the elderly. Am J Clin Nutr 2000;71:614S-620S.
crossref pmid
215. Kado DM, Karlamangla AS, Huang MH, Troen A, Rowe JW, Selhub J, et al. Homocysteine versus the vitamins folate, B6, and B12 as predictors of cognitive function and decline in older high-functioning adults: MacArthur studies of successful aging. Am J Med 2005;118:161-167.
crossref pmid
216. Pieters B, Staals J, Knottnerus I, Rouhl R, Menheere P, Kessels A, et al. Periventricular white matter lucencies relate to low vitamin B12 levels in patients with small vessel stroke. Stroke 2009;40:1623-1626.
crossref pmid
217. de Lau LM, Smith AD, Refsum H, Johnston C, Breteler MM. Plasma vitamin B12 status and cerebral white-matter lesions. J Neurol Neurosurg Psychiatry 2009;80:149-157.
crossref pmid
218. van Overbeek EC, Staals J, van Oostenbrugge RJ. Vitamin B12 and progression of white matter lesions. A 2-year follow-up study in first-ever lacunar stroke patients. PLoS One 2013;8:e78100.
crossref pmid pmc
219. Iosifescu DV, Papakostas GI, Lyoo IK, Lee HK, Renshaw PF, Alpert JE, et al. Brain MRI white matter hyperintensities and one-carbon cycle metabolism in non-geriatric outpatients with major depressive disorder (part I). Psychiatry Res 2005;140:291-299.
crossref pmid
220. Scott TM, Tucker KL, Bhadelia A, Benjamin B, Patz S, Bhadelia R, et al. Homocysteine and B vitamins relate to brain volume and white-matter changes in geriatric patients with psychiatric disorders. Am J Geriatr Psychiatry 2004;12:631-638.
crossref pmid
221. Hickie I, Naismith S, Ward PB, Scott E, Mitchell P, Wilhelm K, et al. Vascular risk and low serum B12 predict white matter lesions in patients with major depression. J Affect Disord 2005;85:327-332.
crossref pmid
222. Tangney CC, Aggarwal NT, Li H, Wilson RS, Decarli C, Evans DA, et al. Vitamin B12, cognition, and brain MRI measures: a cross-sectional examination. Neurology 2011;77:1276-1282.
crossref pmid pmc
223. Narayan SK, Firbank MJ, Saxby BK, Stansby G, Hansrani M, O’Brien JT, et al. Elevated plasma homocysteine is associated with increased brain atrophy rates in older subjects with mild hypertension. Dement Geriatr Cogn Disord 2011;31:341-348.
crossref pmid pdf
224. Sponne IE, Gaire D, Stabler SP, Droesch S, Barbé FM, Allen RH, et al. Inhibition of vitamin B12 metabolism by OH-cobalamin c-lactam in rat oligodendrocytes in culture: a model for studying neuropathy due to vitamin B12 deficiency. Neurosci Lett 2000;288:191-194.
crossref pmid
225. Kim S, Lim IK, Park GH, Paik WK. Biological methylation of myelin basic protein: enzymology and biological significance. Int J Biochem Cell Biol 1997;29:743-751.
crossref pmid
226. Annweiler C, Allali G, Allain P, Bridenbaugh S, Schott AM, Kressig RW, et al. Vitamin D and cognitive performance in adults: a systematic review. Eur J Neurol 2009;16:1083-1089.
crossref pmid
227. Sultan S. Neuroimaging changes associated with vitamin D deficiency-a narrative review. Nutr Neurosci 2022;25:1650-1658.
crossref pmid
228. Zhao Y, Xu J, Feng Z, Wang J. Impact of 25-hydroxy vitamin D on white matter hyperintensity in elderly patients: a systematic review and meta-analysis. Front Neurol 2022;12:721427.
crossref pmid pmc
229. Buell JS, Dawson-Hughes B, Scott TM, Weiner DE, Dallal GE, Qui WQ, et al. 25-hydroxyvitamin D, dementia, and cerebrovascular pathology in elders receiving home services. Neurology 2010;74:18-26.
crossref pmid pmc
230. Prager JM, Thomas C, Ankenbrandt WJ, Meyer JR, Gao Y, Ragin A, et al. Association of white matter hyperintensities with low serum 25-hydroxyvitamin D levels. AJNR Am J Neuroradiol 2014;35:1145-1149.
crossref pmid pmc
231. Chung PW, Park KY, Kim JM, Shin DW, Park MS, Chung YJ, et al. 25-hydroxyvitamin D status is associated with chronic cerebral small vessel disease. Stroke 2015;46:248-251.
crossref pmid
232. Feng C, Tang N, Huang H, Zhang G, Qi X, Shi F. 25-Hydroxy vitamin D level is associated with total MRI burden of cerebral small vessel disease in ischemic stroke patients. Int J Neurosci 2019;129:49-54.
crossref pmid
233. Annweiler C, Bartha R, Karras SN, Gautier J, Roche F, Beauchet O. Vitamin D and white matter abnormalities in older adults: a quantitative volumetric analysis of brain MRI. Exp Gerontol 2015;63:41-47.
crossref pmid
234. Wang L, Zhao XM, Yuan XZ, Wang FY, Shen J, Wang Y. Association between serum 25-hydroxyvitamin D level and cognitive impairment in patients with white matter lesions: a cross-sectional study. Med Princ Pract 2020;29:451-457.
crossref pmid pmc pdf
235. Sakurai T, Ogama N, Toba K. Lower vitamin D is associated with white matter hyperintensity in elderly women with Alzheimer’s disease and amnestic mild cognitive impairment. J Am Geriatr Soc 2014;62:1993-1994.
crossref pmid pdf
236. Schramm S, Schliephake L, Himpfen H, Caspers S, Erbel R, Jöckel KH, et al. Vitamin D and white matter hyperintensities: results of the population-based Heinz Nixdorf Recall Study and 1000BRAINS. Eur J Neurol 2021;28:1849-1858.
crossref pmid pdf
237. Annweiler C, Annweiler T, Bartha R, Herrmann FR, Camicioli R, Beauchet O. Vitamin D and white matter abnormalities in older adults: a cross-sectional neuroimaging study. Eur J Neurol 2014;21:1436-e95.
pmid
238. Michos ED, Carson KA, Schneider AL, Lutsey PL, Xing L, Sharrett AR, et al. Vitamin D and subclinical cerebrovascular disease: the atherosclerosis risk in communities brain magnetic resonance imaging study. JAMA Neurol 2014;71:863-871.
crossref pmid pmc
239. Littlejohns TJ, Kos K, Henley WE, Lang IA, Annweiler C, Beauchet O, et al. Vitamin D and risk of neuroimaging abnormalities. PLoS One 2016;11:e0154896.
crossref pmid pmc
240. Karakis I, Pase MP, Beiser A, Booth SL, Jacques PF, Rogers G, et al. Association of serum vitamin D with the risk of incident dementia and subclinical indices of brain aging: the Framingham heart study. J Alzheimers Dis 2016;51:451-461.
crossref pmid pmc
241. Bowman GL, Silbert LC, Howieson D, Dodge HH, Traber MG, Frei B, et al. Nutrient biomarker patterns, cognitive function, and MRI measures of brain aging. Neurology 2012;78:241-249.
crossref pmid pmc
242. Brouwer-Brolsma EM, van der Zwaluw NL, van Wijngaarden JP, Dhonukshe-Rutten RA, in’t Veld PH, Feskens EJ, et al. Higher serum 25-hydroxyvitamin D and lower plasma glucose are associated with larger gray matter volume but not with white matter or total brain volume in Dutch community-dwelling older adults. J Nutr 2015;145:1817-1823.
crossref pmid
243. Putaala J, Kurkinen M, Tarvos V, Salonen O, Kaste M, Tatlisumak T. Silent brain infarcts and leukoaraiosis in young adults with first-ever ischemic stroke. Neurology 2009;72:1823-1829.
crossref pmid
244. Kim KW, Seo H, Kwak MS, Kim D. Visceral obesity is associated with white matter hyperintensity and lacunar infarct. Int J Obes (Lond) 2017;41:683-688.
crossref pmid pdf
245. Lampe L, Zhang R, Beyer F, Huhn S, Kharabian Masouleh S, Preusser S, et al. Visceral obesity relates to deep white matter hyperintensities via inflammation. Ann Neurol 2019;85:194-203.
crossref pmid pmc pdf
246. Alqarni A, Jiang J, Crawford JD, Koch F, Brodaty H, Sachdev P, et al. Sex differences in risk factors for white matter hyperintensities in non-demented older individuals. Neurobiol Aging 2021;98:197-204.
crossref pmid
247. Seixas AA, Turner AD, Bubu OM, Jean-Louis G, de Leon MJ, Osorio RS, et al. Obesity and race may explain differential burden of white matter hyperintensity load. Clin Interv Aging 2021;16:1563-1571.
crossref pmid pmc pdf
248. Murray AD, McNeil CJ, Salarirad S, Whalley LJ, Staff RT. Early life socioeconomic circumstance and late life brain hyperintensities--a population based cohort study. PLoS One 2014;9:e88969.
crossref pmid pmc
249. Mortamais M, Portet F, Brickman AM, Provenzano FA, Muraskin J, Akbaraly TN, et al. Education modulates the impact of white matter lesions on the risk of mild cognitive impairment and dementia. Am J Geriatr Psychiatry 2014;22:1336-1345.
crossref pmid pmc
250. Schretlen DJ, Inscore AB, Vannorsdall TD, Kraut M, Pearlson GD, Gordon B, et al. Serum uric acid and brain ischemia in normal elderly adults. Neurology 2007;69:1418-1423.
crossref pmid
251. Maniwa K, Yano S, Sheikh AM, Onoda K, Mitaki S, Isomura M, et al. Association between cystatin C gene polymorphism and the prevalence of white matter lesion in elderly healthy subjects. Sci Rep 2020;10:4688.
crossref pmid pmc pdf
252. Backhouse EV, McHutchison CA, Cvoro V, Shenkin SD, Wardlaw JM. Early life risk factors for cerebrovascular disease: a systematic review and meta-analysis. Neurology 2017;88:976-984.
crossref pmid
253. Christensen H, Batterham PJ, Mackinnon AJ, Anstey KJ, Wen W, Sachdev PS. Education, atrophy, and cognitive change in an epidemiological sample in early old age. Am J Geriatr Psychiatry 2009;17:218-226.
crossref pmid
254. Elkins JS, Longstreth WT Jr, Manolio TA, Newman AB, Bhadelia RA, Johnston SC. Education and the cognitive decline associated with MRI-defined brain infarct. Neurology 2006;67:435-440.
crossref pmid
255. Breteler MM, van Swieten JC, Bots ML, Grobbee DE, Claus JJ, van den Hout JH, et al. Cerebral white matter lesions, vascular risk factors, and cognitive function in a population-based study: the Rotterdam study. Neurology 1994;44:1246-1252.
crossref pmid
256. Habes M, Sotiras A, Erus G, Toledo JB, Janowitz D, Wolk DA, et al. White matter lesions: spatial heterogeneity, links to risk factors, cognition, genetics, and atrophy. Neurology 2018;91:e964-e975.
pmid pmc
257. Sachdev PS, Parslow R, Wen W, Anstey KJ, Easteal S. Sex differences in the causes and consequences of white matter hyperintensities. Neurobiol Aging 2009;30:946-956.
crossref pmid
258. Sullivan P, Pary R, Telang F, Rifai AH, Zubenko GS. Risk factors for white matter changes detected by magnetic resonance imaging in the elderly. Stroke 1990;21:1424-1428.
crossref pmid
259. Schmidt R, Fazekas F, Kleinert G, Offenbacher H, Gindl K, Payer F, et al. Magnetic resonance imaging signal hyperintensities in the deep and subcortical white matter. A comparative study between stroke patients and normal volunteers. Arch Neurol 1992;49:825-827.
crossref pmid
260. Assareh AA, Mather KA, Crawford JD, Wen W, Anstey KJ, Easteal S, et al. Renin-angiotensin system genetic polymorphisms and brain white matter lesions in older Australians. Am J Hypertens 2014;27:1191-1198.
crossref pmid
261. Geerlings MI, Appelman AP, Vincken KL, Algra A, Witkamp TD, Mali WP, et al. Brain volumes and cerebrovascular lesions on MRI in patients with atherosclerotic disease. The SMARTMR study. Atherosclerosis 2010;210:130-136.
crossref pmid
262. Brickman AM, Schupf N, Manly JJ, Luchsinger JA, Andrews H, Tang MX, et al. Brain morphology in older African Americans, Caribbean Hispanics, and whites from northern Manhattan. Arch Neurol 2008;65:1053-1061.
crossref pmid pmc
263. Nyquist PA, Bilgel MS, Gottesman R, Yanek LR, Moy TF, Becker LC, et al. Extreme deep white matter hyperintensity volumes are associated with African American race. Cerebrovasc Dis 2014;37:244-250.
crossref pmid pmc pdf
264. Gottesman RF, Coresh J, Catellier DJ, Sharrett AR, Rose KM, Coker LH, et al. Blood pressure and white-matter disease progression in a biethnic cohort: atherosclerosis risk in communities (ARIC) study. Stroke 2010;41:3-8.
crossref pmid pmc
265. Turner ST, Jack CR, Fornage M, Mosley TH, Boerwinkle E, de Andrade M. Heritability of leukoaraiosis in hypertensive sibships. Hypertension 2004;43:483-487.
crossref pmid
266. Atwood LD, Wolf PA, Heard-Costa NL, Massaro JM, Beiser A, D’Agostino RB, et al. Genetic variation in white matter hyperintensity volume in the Framingham study. Stroke 2004;35:1609-1613.
crossref pmid
267. Sachdev PS, Thalamuthu A, Mather KA, Ames D, Wright MJ, Wen W; OATS Collaborative Research Team. White matter hyperintensities are under strong genetic influence. Stroke 2016;47:1422-1428.
crossref pmid
268. Paternoster L, Chen W, Sudlow CL. Genetic determinants of white matter hyperintensities on brain scans: a systematic assessment of 19 candidate gene polymorphisms in 46 studies in 19,000 subjects. Stroke 2009;40:2020-2026.
crossref pmid
269. Lopez LM, Hill WD, Harris SE, Valdes Hernandez M, Munoz Maniega S, Bastin ME, et al. Genes from a translational analysis support a multifactorial nature of white matter hyperintensities. Stroke 2015;46:341-347.
crossref pmid pmc
270. Jian X, Satizabal CL, Smith AV, Wittfeld K, Bis JC, Smith JA, et al. Exome chip analysis identifies low-frequency and rare variants in MRPL38 for white matter hyperintensities on brain magnetic resonance imaging. Stroke 2018;49:1812-1819.
pmid pmc
271. Yadav BK, Shin BS. Single-nucleotide polymorphisms of tight junction component claudin-1 associated with leukoaraiosis. J Stroke Cerebrovasc Dis 2015;24:1662-1670.
crossref pmid
272. Oliveira-Filho J, Ornellas AC, Zhang CR, Oliveira LM, AraújoSantos T, Borges VM, et al. COX-2 rs20417 polymorphism is associated with stroke and white matter disease. J Stroke Cerebrovasc Dis 2015;24:1817-1822.
crossref pmid pmc
273. Zhang M, Zhu W, Yun W, Wang Q, Cheng M, Zhang Z, et al. Correlation of matrix metalloproteinase-2 single nucleotide polymorphisms with the risk of small vessel disease (SVD). J Neurol Sci 2015;356:61-64.
crossref pmid
274. Yadav BK, Oh SY, Kim NK, Shin BS. Association of rs2075575 and rs9951307 polymorphisms of AQP-4 gene with leukoaraiosis. J Stroke Cerebrovasc Dis 2014;23:1199-1206.
crossref pmid
275. Huang WQ, Ye HM, Li FF, Yi KH, Zhang Y, Cai LL, et al. Analysis of genetic polymorphisms associated with leukoaraiosis in the southern Chinese population: a case-control study. Medicine (Baltimore) 2016;95:e3857.

276. Huang WQ, Ye HM, Cai LL, Ma QL, Lu CX, Tong SJ, et al. The associations of PMF1, ICAM1, AGT, TRIM65, FBF1, and ACOX1 variants with leukoaraiosis in Chinese population. Front Genet 2019;10:615.
crossref pmid pmc
277. Yadav BK, Shin BS. Single-nucleotide polymorphisms of the adherent junction component cadherin gene are associated with leukoaraiosis. Gene 2018;676:65-72.
crossref pmid
278. Yadav BK, Yadav R, Kang HG, Kim KW, Lee CH, Shin BS. Association of genetic variation in a Wnt signaling pathway gene (β-catenin) with susceptibility to leukoaraiosis. Genet Test Mol Biomarkers 2020;24:708-716.
crossref pmid
279. Li J, Abedi V, Zand R, Griessenauer CJ. Replication of top loci from COL4A1/2 associated with white matter hyperintensity burden in patients with ischemic stroke. Stroke 2020;51:3751-3755.
crossref pmid pmc
280. Parikh NS, Dueker N, Varela D, Del Brutto VJ, Rundek T, Wright CB, et al. Association between PNPLA3 rs738409 G variant and MRI cerebrovascular disease biomarkers. J Neurol Sci 2020;416:116981.
crossref pmid pmc
281. Davis CM, Bah TM, Zhang WH, Nelson JW, Golgotiu K, Nie X, et al. GPR39 localization in the aging human brain and correlation of expression and polymorphism with vascular cognitive impairment. Alzheimers Dement (N Y) 2021;7:e12214.
crossref pmid pmc pdf
282. Liu JY, Yao M, Dai Y, Han F, Zhai FF, Zhang DD, et al. Rare NOTCH3 variants in a Chinese population-based cohort and its relationship with cerebral small vessel disease. Stroke 2021;52:3918-3925.
crossref pmid
283. Ferroni P, Palmirotta R, Egeo G, Aurilia C, Valente MG, Spila A, et al. Association of LTA and SOD gene polymorphisms with cerebral white matter hyperintensities in migraine patients. Int J Mol Sci 2022;23:13781.
crossref pmid pmc
284. Gao Y, Su B, Luo Y, Tian Y, Hong S, Gao S, et al. HLA-C*07:01 and HLA-DQB1*02:01 protect against white matter hyperintensities and deterioration of cognitive function: a population-based cohort study. Brain Behav Immun 2024;115:250-257.
crossref pmid
285. Fornage M, Debette S, Bis JC, Schmidt H, Ikram MA, Dufouil C, et al. Genome-wide association studies of cerebral white matter lesion burden: the CHARGE consortium. Ann Neurol 2011;69:928-939.
pmid pmc
286. Verhaaren BF, Debette S, Bis JC, Smith JA, Ikram MK, Adams HH, et al. Multiethnic genome-wide association study of cerebral white matter hyperintensities on MRI. Circ Cardiovasc Genet 2015;8:398-409.
pmid pmc
287. Malik R, Beaufort N, Frerich S, Gesierich B, Georgakis MK, Rannikmäe K, et al. Whole-exome sequencing reveals a role of HTRA1 and EGFL8 in brain white matter hyperintensities. Brain 2021;144:2670-2682.
crossref pmid pmc pdf
288. Traylor M, Tozer DJ, Croall ID, Lisiecka-Ford DM, Olorunda AO, Boncoraglio G, et al. Genetic variation in PLEKHG1 is associated with white matter hyperintensities (n = 11,226). Neurology 2019;92:e749-e757.
crossref pmid pmc
289. Persyn E, Hanscombe KB, Howson JMM, Lewis CM, Traylor M, Markus HS. Genome-wide association study of MRI markers of cerebral small vessel disease in 42,310 participants. Nat Commun 2020;11:2175.
crossref pmid pmc pdf
290. Rutten-Jacobs LCA, Tozer DJ, Duering M, Malik R, Dichgans M, Markus HS, et al. Genetic study of white matter integrity in UK Biobank (n=8448) and the overlap with stroke, depression, and dementia. Stroke 2018;49:1340-1347.
crossref pmid pmc
291. Armstrong NJ, Mather KA, Sargurupremraj M, Knol MJ, Malik R, Satizabal CL, et al. Common genetic variation indicates separate causes for periventricular and deep white matter hyperintensities. Stroke 2020;51:2111-2121.
pmid pmc
292. Simpson JE, Hosny O, Wharton SB, Heath PR, Holden H, Fernando MS, et al. Microarray RNA expression analysis of cerebral white matter lesions reveals changes in multiple functional pathways. Stroke 2009;40:369-375.
crossref pmid
293. Xu H, Stamova B, Jickling G, Tian Y, Zhan X, Ander BP, et al. Distinctive RNA expression profiles in blood associated with white matter hyperintensities in brain. Stroke 2010;41:2744-2749.
crossref pmid pmc
294. Lin H, Satizabal C, Xie Z, Yang Q, Huan T, Joehanes R, et al. Whole blood gene expression and white matter hyperintensities. Mol Neurodegener 2017;12:67.
crossref pmid pmc pdf
295. Jickling GC, Ander BP, Zhan X, Stamova B, Hull H, DeCarli C, et al. Progression of cerebral white matter hyperintensities is related to leucocyte gene expression. Brain 2022;145:3179-3186.
crossref pmid pmc pdf
296. Hou XH, Bi YL, Tan MS, Xu W, Li JQ, Shen XN, et al. Genomewide association study identifies Alzheimer’s risk variant in MS4A6A influencing cerebrospinal fluid sTREM2 levels. Neurobiol Aging 2019;84:241.e13-241.e20.
crossref pmid
297. Peters XQ, Malinga TH, Agoni C, Olotu FA, Soliman MES. Zoning in on Tankyrases: a brief review on the past, present and prospective studies. Anticancer Agents Med Chem 2019;19:1920-1934.
crossref pmid
298. Yang HY, Shen JX, Wang Y, Liu Y, Shen DY, Quan S. Tankyrase promotes aerobic glycolysis and proliferation of ovarian cancer through activation of Wnt/β-catenin signaling. Biomed Res Int 2019;2019:2686340.
crossref pmid pmc pdf
299. Iwaya T, Maesawa C, Kimura T, Ogasawara S, Ikeda K, Kimura Y, et al. Infrequent mutation of the human envoplakin gene is closely linked to the tylosis oesophageal cancer locus in sporadic oesophageal squamous cell carcinomas. Oncol Rep 2005;13:703-707.
crossref pmid
300. Weber R, Weimar C, Blatchford J, Hermansson K, Wanke I, Möller-Hartmann C, et al. Telmisartan on top of antihypertensive treatment does not prevent progression of cerebral white matter lesions in the prevention regimen for effectively avoiding second strokes (PRoFESS) MRI substudy. Stroke 2012;43:2336-2342.
crossref pmid
301. Peng J, Lu F, Wang Z, Zhong M, Sun L, Hu N, et al. Excessive lowering of blood pressure is not beneficial for progression of brain white matter hyperintensive and cognitive impairment in elderly hypertensive patients: 4-year follow-up study. J Am Med Dir Assoc 2014;15:904-910.
crossref pmid
302. Murray AM, Hsu FC, Williamson JD, Bryan RN, Gerstein HC, Sullivan MD, et al. ACCORDION MIND: results of the observational extension of the ACCORD MIND randomised trial. Diabetologia 2017;60:69-80.
crossref pmid pmc pdf
303. Godin O, Tzourio C, Maillard P, Mazoyer B, Dufouil C. Antihypertensive treatment and change in blood pressure are associated with the progression of white matter lesion volumes: the three-city (3C)-Dijon magnetic resonance imaging study. Circulation 2011;123:266-273.
crossref pmid
304. Schiffrin EL. Blood pressure lowering in PROGRESS (perindopril protection against recurrent stroke study) and white matter hyperintensities: should this progress matter to patients? Circulation 2005;112:1525-1526.
crossref pmid
305. van Dalen JW, Moll van Charante EP, Caan MWA, Scheltens P, Majoie CBLM, Nederveen AJ, et al. Effect of long-term vascular care on progression of cerebrovascular lesions: magnetic resonance imaging substudy of the PreDIVA trial (prevention of dementia by intensive vascular care). Stroke 2017;48:1842-1848.
crossref pmid
306. de Havenon A, Majersik JJ, Tirschwell DL, McNally JS, Stoddard G, Rost NS. Blood pressure, glycemic control, and white matter hyperintensity progression in type 2 diabetics. Neurology 2019;92:e1168-e1175.
pmid pmc
307. White WB, Wakefield DB, Moscufo N, Guttmann CRG, Kaplan RF, Bohannon RW, et al. Effects of intensive versus standard ambulatory blood pressure control on cerebrovascular outcomes in older people (INFINITY). Circulation 2019;140:1626-1635.
crossref pmid pmc
308. Lai Y, Jiang C, Du X, Sang C, Guo X, Bai R, et al. Effect of intensive blood pressure control on the prevention of white matter hyperintensity: systematic review and meta-analysis of randomized trials. J Clin Hypertens (Greenwich) 2020;22:1968-1973.
crossref pmid pmc pdf
309. Su C, Wu H, Yang X, Zhao B, Zhao R. The relation between antihypertensive treatment and progression of cerebral small vessel disease: a systematic review and meta-analysis of randomized controlled trials. Medicine (Baltimore) 2021;100:e26749.
pmid pmc
310. Heutz RA, Weijs RW, de Heus RA, Claassen JA. Antihypertensives in dementia: good or bad for the brain? J Cereb Blood Flow Metab 2023;43:1796-1799.
crossref pmid pmc pdf
311. Willmot M, Ghadami A, Whysall B, Clarke W, Wardlaw J, Bath PM. Transdermal glyceryl trinitrate lowers blood pressure and maintains cerebral blood flow in recent stroke. Hypertension 2006;47:1209-1215.
crossref pmid
312. Sare GM, Gray LJ, Bath PM. Effect of antihypertensive agents on cerebral blood flow and flow velocity in acute ischaemic stroke: systematic review of controlled studies. J Hypertens 2008;26:1058-1064.
crossref pmid
313. Kate M, Asdaghi N, Gioia LC, Buck B, Majumdar SR, Jeerakathil T, et al. Blood pressure reduction in hypertensive acute ischemic stroke patients does not affect cerebral blood flow. J Cereb Blood Flow Metab 2019;39:1878-1887.
crossref pmid pmc pdf
314. Croall ID, Tozer DJ, Moynihan B, Khan U, O’Brien JT, Morris RG, et al. Effect of standard vs intensive blood pressure control on cerebral blood flow in small vessel disease: the PRESERVE randomized clinical trial. JAMA Neurol 2018;75:720-727.
crossref pmid pmc
315. de Heus RAA, de Jong DLK, Lawlor BL, Claassen JAHR; NILVAD Study Group. Longitudinal changes in the control mechanisms for blood pressure and cerebral blood flow in Alzheimer’s disease: secondary results of a randomized controlled trial. Cereb Circ Cogn Behav 2021;2:100024.
crossref pmid pmc
316. van Rijssel AE, Stins BC, Beishon LC, Sanders ML, Quinn TJ, Claassen JAHR, et al. Effect of antihypertensive treatment on cerebral blood flow in older adults: a systematic review and meta-analysis. Hypertension 2022;79:1067-1078.
crossref pmid pmc
317. Efimova NY, Chernov VI, Efimova IY, Lishmanov YB. Influence of antihypertensive therapy on cerebral perfusion in patients with metabolic syndrome: relationship with cognitive function and 24-h arterial blood pressure monitoring. Cardiovasc Ther 2015;33:209-215.
crossref pmid
318. Dolui S, Detre JA, Gaussoin SA, Herrick JS, Wang DJJ, Tamura MK, et al. Association of intensive vs standard blood pressure control with cerebral blood flow: secondary analysis of the SPRINT MIND randomized clinical trial. JAMA Neurol 2022;79:380-389.
crossref pmid pmc
319. Ikeme JC, Pergola PE, Scherzer R, Shlipak MG, Catanese L, McClure LA, et al. Cerebral white matter hyperintensities, kidney function decline, and recurrent stroke after intensive blood pressure lowering: results from the secondary prevention of small subcortical strokes (SPS3) trial. J Am Heart Assoc 2019;8:e010091.
crossref pmid pmc
320. Williamson JD, Launer LJ, Bryan RN, Coker LH, Lazar RM, Gerstein HC, et al. Cognitive function and brain structure in persons with type 2 diabetes mellitus after intensive lowering of blood pressure and lipid levels: a randomized clinical trial. JAMA Intern Med 2014;174:324-333.
crossref pmid pmc
321. Verghese J, Lipton RB, Hall CB, Kuslansky G, Katz MJ. Low blood pressure and the risk of dementia in very old individuals. Neurology 2003;61:1667-1672.
crossref pmid
322. McGrath ER, Beiser AS, DeCarli C, Plourde KL, Vasan RS, Greenberg SM, et al. Blood pressure from mid- to late life and risk of incident dementia. Neurology 2017;89:2447-2454.
crossref pmid pmc
323. Yeung A, Kiss A, Gallagher D. Intensive control of hypertension and risk of Alzheimer’s dementia in older adults with depression. Int J Geriatr Psychiatry 2020;35:888-896.
crossref pmid pmc pdf
324. Jiang C, Lai Y, Du X, Wang Y, Li S, He L, et al. Effects of intensive blood pressure control on cardiovascular and cognitive outcomes in patients with atrial fibrillation: insights from the SPRINT trial. Europace 2022;24:1560-1568.
crossref pmid pdf
325. Poon IO. Effects of antihypertensive drug treatment on the risk of dementia and cognitive impairment. Pharmacotherapy 2008;28:366-375.
crossref pmid
326. Hughes D, Judge C, Murphy R, Loughlin E, Costello M, Whiteley W, et al. Association of blood pressure lowering with incident dementia or cognitive impairment: a systematic review and meta-analysis. JAMA 2029;323:1934-1944.
crossref pmid pmc
327. Peters R, Xu Y, Fitzgerald O, Aung HL, Beckett N, Bulpitt C, et al. Blood pressure lowering and prevention of dementia: an individual patient data meta-analysis. Eur Heart J 2022;43:4980-4990.
crossref pmid pdf
328. Ding J, Davis-Plourde KL, Sedaghat S, Tully PJ, Wang W, Phillips C, et al. Antihypertensive medications and risk for incident dementia and Alzheimer’s disease: a meta-analysis of individual participant data from prospective cohort studies. Lancet Neurol 2020;19:61-70.
crossref pmid
329. Lee CJ, Hwang J, Kang CY, Kim HC, Ryu DR, Ihm SH, et al. Protective effect of controlled blood pressure on risk of dementia in low-risk, grade 1 hypertension. J Hypertens 2021;39:1662-1669.
crossref pmid
330. Canavan M, O’Donnell MJ. Hypertension and cognitive impairment: a review of mechanisms and key concepts. Front Neurol 2022;13:821135.
crossref pmid pmc
331. Tully PJ, Dartigues JF, Debette S, Helmer C, Artero S, Tzourio C. Dementia risk with antihypertensive use and blood pressure variability: a cohort study. Neurology 2016;87:601-608.
crossref pmid
332. Lennon MJ, Lam BCP, Lipnicki DM, Crawford JD, Peters R, Schutte AE, et al. Use of antihypertensives, blood pressure, and estimated risk of dementia in late life: an individual participant data meta-analysis. JAMA Netw Open 2023;6:e2333353.
crossref pmid pmc
333. Jiang C, Li S, Wang Y, Lai Y, Bai Y, Zhao M, et al. Diastolic blood pressure and intensive blood pressure control on cognitive outcomes: insights from the SPRINT MIND trial. Hypertension 2023;80:580-589.
crossref pmid
334. Peters R, Beckett N, Forette F, Tuomilehto J, Clarke R, Ritchie C, et al. Incident dementia and blood pressure lowering in the hypertension in the very elderly trial cognitive function assessment (HYVET-COG): a double-blind, placebo controlled trial. Lancet Neurol 2008;7:683-689.
crossref pmid
335. van Middelaar T, van Vught LA, van Gool WA, Simons EMF, van den Born BH, Moll van Charante EP, et al. Blood pressurelowering interventions to prevent dementia: a systematic review and meta-analysis. J Hypertens 2018;36:1780-1787.
pmid
336. SPRINT MIND Investigators for the SPRINT Research Group. Effect of intensive vs standard blood pressure control on probable dementia: a randomized clinical trial. JAMA 2019;321:553-561.
pmid pmc
337. Rojas-Saunero LP, Hilal S, Murray EJ, Logan RW, Ikram MA, Swanson SA. Hypothetical blood-pressure-lowering interventions and risk of stroke and dementia. Eur J Epidemiol 2021;36:69-79.
crossref pmid pmc pdf
338. Kellar D, Lockhart SN, Aisen P, Raman R, Rissman RA, Brewer J, et al. Intranasal insulin reduces white matter hyperintensity progression in association with improvements in cognition and CSF biomarker profiles in mild cognitive impairment and Alzheimer’s disease. J Prev Alzheimers Dis 2021;8:240-248.
crossref pmid pmc
339. Inkeri J, Adeshara K, Harjutsalo V, Forsblom C, Liebkind R, Tatlisumak T, et al. Glycemic control is not related to cerebral small vessel disease in neurologically asymptomatic individuals with type 1 diabetes. Acta Diabetol 2022;59:481-490.
crossref pmid pmc pdf
340. Livny A, Ravona-Springer R, Heymann A, Priess R, Kushnir T, Tsarfaty G, et al. Long-term variability in glycemic control is associated with white matter hyperintensities in APOE4 genotype carriers with type 2 diabetes. Diabetes Care 2016;39:1056-1059.
crossref pmid pmc pdf
341. Launer LJ, Miller ME, Williamson JD, Lazar RM, Gerstein HC, Murray AM, et al. Effects of intensive glucose lowering on brain structure and function in people with type 2 diabetes (ACCORD MIND): a randomised open-label substudy. Lancet Neurol 2011;10:969-977.
crossref pmid pmc
342. Wardlaw JM, Debette S, Jokinen H, De Leeuw FE, Pantoni L, Chabriat H, et al. ESO guideline on covert cerebral small vessel disease. Eur Stroke J 2021;6:CXI-CLXII.
crossref pmid pmc pdf
343. Mortensen MB, Falk E. Primary prevention with statins in the elderly. J Am Coll Cardiol 2018;71:85-94.
crossref pmid
344. Cholesterol Treatment Trialists’ Collaboration. Efficacy and safety of statin therapy in older people: a meta-analysis of individual participant data from 28 randomised controlled trials. Lancet 2019;393:407-415.
pmid pmc
345. Martínez-Sánchez P, Rivera-Ordóñez C, Fuentes B, Ortega-Casarrubios MA, Idrovo L, Díez-Tejedor E. The beneficial effect of statins treatment by stroke subtype. Eur J Neurol 2009;16:127-133.
crossref pmid
346. Bernick C, Katz R, Smith NL, Rapp S, Bhadelia R, Carlson M, et al. Statins and cognitive function in the elderly: the cardiovascular health study. Neurology 2005;65:1388-1394.
crossref pmid
347. ten Dam VH, van den Heuvel DM, van Buchem MA, Westendorp RG, Bollen EL, Ford I, et al. Effect of pravastatin on cerebral infarcts and white matter lesions. Neurology 2005;64:1807-1809.
crossref pmid
348. Ramanan VK, Przybelski SA, Graff-Radford J, Castillo AM, Lowe VJ, Mielke MM, et al. Statins and brain health: Alzheimer’s disease and cerebrovascular disease biomarkers in older adults. J Alzheimers Dis 2018;65:1345-1352.
crossref pmid pmc
349. Vogt NM, Hunt JFV, Ma Y, Van Hulle CA, Adluru N, Chappell RJ, et al. Effects of simvastatin on white matter integrity in healthy middle-aged adults. Ann Clin Transl Neurol 2021;8:1656-1667.
crossref pmid pmc pdf
350. Mok VC, Lam WW, Fan YH, Wong A, Ng PW, Tsoi TH, et al. Effects of statins on the progression of cerebral white matter lesion: post hoc analysis of the ROCAS (regression of cerebral artery stenosis) study. J Neurol 2009;256:750-757.
crossref pmid pdf
351. Ji T, Zhao Y, Wang J, Cui Y, Duan D, Chai Q, et al. Effect of low-dose statins and apolipoprotein E genotype on cerebral small vessel disease in older hypertensive patients: a subgroup analysis of a randomized clinical trial. J Am Med Dir Assoc 2018;19:995-1002.e4.
crossref pmid
352. Zhang H, Cui Y, Zhao Y, Dong Y, Duan D, Wang J, et al. Effects of sartans and low-dose statins on cerebral white matter hyperintensities and cognitive function in older patients with hypertension: a randomized, double-blind and placebocontrolled clinical trial. Hypertens Res 2019;42:717-729.
crossref pmid pdf
353. Guo Y, Li Y, Liu X, Cui Y, Zhao Y, Sun S, et al. Assessing the effectiveness of statin therapy for alleviating cerebral small vessel disease progression in people ≥75years of age. BMC Geriatr 2020;20:292.
crossref pmid pmc pdf
354. Xue J, Wu Z, Gong S, Qin S, Gu A. High-dose atorvastatin improves vascular endothelial function in patients with leukoaraiosis. J Clin Lab Anal 2020;34:e23081.
crossref pmid pmc pdf
355. Sterzer P, Meintzschel F, Rösler A, Lanfermann H, Steinmetz H, Sitzer M. Pravastatin improves cerebral vasomotor reactivity in patients with subcortical small-vessel disease. Stroke 2001;32:2817-2820.
crossref pmid
356. Ii M, Losordo DW. Statins and the endothelium. Vascul Pharmacol 2007;46:1-9.
crossref pmid
357. Liu Z, Zhao Y, Wei F, Ye L, Lu F, Zhang H, et al. Treatment with telmisartan/rosuvastatin combination has a beneficial synergistic effect on ameliorating Th17/Treg functional imbalance in hypertensive patients with carotid atherosclerosis. Atherosclerosis 2014;233:291-299.
crossref pmid
358. Janic´ M, Lunder M, Šabovicˇ M. A low-dose combination of fluvastatin and valsartan: a new “drug” and a new approach for decreasing the arterial age. Biomed Res Int 2015;2015:235709.
pmid pmc
359. Rizos CV, Liberopoulos EN, Tellis CC, Tselepis AD, Elisaf MS. The effect of combining rosuvastatin with sartans of different peroxisome proliferator receptor-γ activating capacity on plasma 8-isoprostane prostaglandin F2a levels. Arch Med Sci 2013;9:172-176.
crossref pmid pmc
360. Kim JS. Role of blood lipid levels and lipid-lowering therapy in stroke patients with different levels of cerebral artery diseases: reconsidering recent stroke guidelines. J Stroke 2021;23:149-161.
crossref pmid pmc pdf
361. Collins R, Armitage J, Parish S, Sleight P, Peto R; Heart Protection Study Collaborative Group. Effects of cholesterollowering with simvastatin on stroke and other major vascular events in 20536 people with cerebrovascular disease or other high-risk conditions. Lancet 2004;363:757-767.
crossref pmid
362. Goldstein LB, Amarenco P, Szarek M, Callahan A 3rd, Hennerici M, Sillesen H, et al. Hemorrhagic stroke in the stroke prevention by aggressive reduction in cholesterol levels study. Neurology 2008;70(24 Pt 2):2364-2370.
crossref pmid
363. Hosomi N, Nagai Y, Kohriyama T, Ohtsuki T, Aoki S, Nezu T, et al. The Japan statin treatment against recurrent stroke (JSTARS): a multicenter, randomized, open-label, parallel-group study. EBioMedicine 2015;2:1071-1078.
crossref pmid pmc
364. Hosomi N, Kitagawa K, Nagai Y, Nakagawa Y, Aoki S, Nezu T, et al. Different influences of statin treatment in preventing at-risk stroke subtypes: a post hoc analysis of J-STARS. J Atheroscler Thromb 2020;27:449-460.
crossref pmid pmc
365. Boxer AL, Kramer JH, Johnston K, Goldman J, Finley R, Miller BL. Executive dysfunction in hyperhomocystinemia responds to homocysteine-lowering treatment. Neurology 2005;64:1431-1434.
crossref pmid
366. Cavalieri M, Schmidt R, Chen C, Mok V, de Freitas GR, Song S, et al. B vitamins and magnetic resonance imaging-detected ischemic brain lesions in patients with recent transient ischemic attack or stroke: the VITAmins TO Prevent Stroke (VITATOPS) MRI-substudy. Stroke 2012;43:3266-3270.
crossref pmid
367. Fassbender K, Mielke O, Bertsch T, Nafe B, Fröschen S, Hennerici M. Homocysteine in cerebral macroangiography and microangiopathy. Lancet 1999;353:1586-1587.
crossref pmid
368. Weir DG, Scott JM. Brain function in the elderly: role of vitamin B12 and folate. Br Med Bull 1999;55:669-682.
crossref pmid
369. Park HK, Kob SB, Jung KH, Jang MU, Kim DH, Kim JT, et al. 2022 Update of the Korean clinical practice guidelines for stroke: antithrombotic therapy for patients with acute ischemic stroke or transient ischemic attack. J Stroke 2022;24:166-175.
crossref pmid pmc pdf
370. Ishida K, Messé SR. Antiplatelet strategies for secondary prevention of stroke and TIA. Curr Atheroscler Rep 2014;16:449.
crossref pmid pdf
371. Ter Telgte A, van Leijsen EMC, Wiegertjes K, Klijn CJM, Tuladhar AM, de Leeuw FE. Cerebral small vessel disease: from a focal to a global perspective. Nat Rev Neurol 2018;14:387-398.
crossref pmid pdf
372. Huang N, Chen D, Wu X, Chen X, Zhang X, Niu J, et al. Aspirin promotes oligodendroglial differentiation through inhibition of Wnt signaling pathway. Mol Neurobiol 2016;53:3258-3266.
crossref pmid pdf
373. Chen J, Zuo S, Wang J, Huang J, Zhang X, Liu Y, et al. Aspirin promotes oligodendrocyte precursor cell proliferation and differentiation after white matter lesion. Front Aging Neurosci 2014;6:7.
crossref pmid pmc
374. Holcombe A, Ammann E, Espeland MA, Kelley BJ, Manson JE, Wallace R, et al. Chronic use of aspirin and total white matter lesion volume: results from the women’s health initiative memory study of magnetic resonance imaging study. J Stroke Cerebrovasc Dis 2017;26:2128-2136.
crossref pmid
375. Ward SA, Raniga P, Ferris NJ, Woods RL, Storey E, Bailey MJ, et al. ASPREE-NEURO study protocol: a randomized controlled trial to determine the effect of low-dose aspirin on cerebral microbleeds, white matter hyperintensities, cognition, and stroke in the healthy elderly. Int J Stroke 2017;12:108-113.
crossref pmid pmc pdf
376. Zheng SL, Roddick AJ. Association of aspirin use for primary prevention with cardiovascular events and bleeding events: a systematic review and meta-analysis. JAMA 2019;321:277-287.
crossref pmid pmc
377. Shinohara Y, Katayama Y, Uchiyama S, Yamaguchi T, Handa S, Matsuoka K, et al. Cilostazol for prevention of secondary stroke (CSPS 2): an aspirin-controlled, double-blind, randomised noninferiority trial. Lancet Neurol 2010;9:959-968.
crossref pmid
378. Lin MP, Meschia JF, Gopal N, Barrett KM, Ross OA, Ertekin-Taner N, et al. Cilostazol versus aspirin for secondary stroke prevention: systematic review and meta-analysis. J Stroke Cerebrovasc Dis 2021;30:105581.
crossref pmid
379. Han HJ, Kim BC, Youn YC, Jeong JH, Kim JH, Lee JH, et al. A comparison study of cilostazol and aspirin on changes in volume of cerebral small vessel disease white matter changes: protocol of a multicenter, randomized controlled trial. Dement Neurocogn Disord 2019;18:138-148.
crossref pmid pmc pdf
380. Kim BC, Youn YC, Jeong JH, Han HJ, Kim JH, Lee JH, et al. Cilostazol versus aspirin on white matter changes in cerebral small vessel disease: a randomized controlled trial. Stroke 2022;53:698-709.
crossref pmid
381. Ip BYM, Lam BYK, Hui VMH, Au LWC, Liu MWT, Shi L, et al. Efficacy and safety of cilostazol in decreasing progression of cerebral white matter hyperintensities—a randomized controlled trial. Alzheimers Dement (N Y) 2022;8:e12369.
crossref pmid pmc pdf
382. Gons RA, van Norden AG, de Laat KF, van Oudheusden LJ, van Uden IW, Zwiers MP, et al. Cigarette smoking is associated with reduced microstructural integrity of cerebral white matter. Brain 2011;134(Pt 7):2116-2124.
crossref pmid
383. Akbar Z, Fituri S, Ouagueni A, Alalwani J, Sukik A, Al-Jayyousi GF, et al. Associations of the MIND diet with cardiometabolic diseases and their risk factors: a systematic review. Diabetes Metab Syndr Obes 2023;16:3353-3371.
crossref pmid pmc pdf
384. Makin SDJ, Mubki GF, Doubal FN, Shuler K, Staals J, Dennis MS, et al. Small vessel disease and dietary salt intake: cross-sectional study and systematic review. J Stroke Cerebrovasc Dis 2017;26:3020-3028.
crossref pmid pmc
385. Strazzullo P, D’Elia L, Kandala NB, Cappuccio FP. Salt intake, stroke, and cardiovascular disease: meta-analysis of prospective studies. BMJ 2009;339:b4567.
crossref pmid pmc
386. Del Brutto OH, Recalde BY, Mera RM. Dietary oily fish intake is inversely associated with severity of white matter hyperintensities of presumed vascular origin. A population-based study in frequent fish consumers of Amerindian ancestry. J Stroke Cerebrovasc Dis 2021;30:105778.
crossref pmid
387. Song S, Gaynor AM, Cruz E, Lee S, Gazes Y, Habeck C, et al. Mediterranean diet and white matter hyperintensity change over time in cognitively intact adults. Nutrients 2022;14:3664.
crossref pmid pmc
388. Barnes LL, Dhana K, Liu X, Carey VJ, Ventrelle J, Johnson K, et al. Trial of the MIND diet for prevention of cognitive decline in older persons. N Engl J Med 2023;389:602-611.
crossref pmid pmc
389. Venkatraman VK, Sanderson A, Cox KL, Ellis KA, Steward C, Phal PM, et al. Effect of a 24-month physical activity program on brain changes in older adults at risk of Alzheimer’s disease: the AIBL active trial. Neurobiol Aging 2020;89:132-141.
crossref pmid
390. Ho AJ, Raji CA, Becker JT, Lopez OL, Kuller LH, Hua X, et al. The effects of physical activity, education, and body mass index on the aging brain. Hum Brain Mapp 2011;32:1371-1382.
crossref pmid pmc
391. Rosano C, Venkatraman VK, Guralnik J, Newman AB, Glynn NW, Launer L, et al. Psychomotor speed and functional brain MRI 2 years after completing a physical activity treatment. J Gerontol A Biol Sci Med Sci 2010;65:639-647.
crossref pmid pmc
392. Willey JZ, Moon YP, Paik MC, Yoshita M, Decarli C, Sacco RL, et al. Lower prevalence of silent brain infarcts in the physically active: the northern Manhattan study. Neurology 2011;76:2112-2118.
crossref pmid pmc
393. Smith JA, Turner ST, Sun YV, Fornage M, Kelly RJ, Mosley TH, et al. Complexity in the genetic architecture of leukoaraiosis in hypertensive sibships from the GENOA study. BMC Med Genomics 2009;2:16.
crossref pmid pmc pdf
394. Sen A, Gider P, Cavalieri M, Freudenberger P, Farzi A, Schallert M, et al. Association of cardiorespiratory fitness and morphological brain changes in the elderly: results of the Austrian stroke prevention study. Neurodegener Dis 2012;10:135-137.
crossref pmid pdf
395. Tseng BY, Gundapuneedi T, Khan MA, Diaz-Arrastia R, Levine BD, Lu H, et al. White matter integrity in physically fit older adults. Neuroimage 2013;82:510-516.
crossref pmid pmc
396. Gow AJ, Bastin ME, Muñoz Maniega S, Valdés Hernández MC, Morris Z, Murray C, et al. Neuroprotective lifestyles and the aging brain: activity, atrophy, and white matter integrity. Neurology 2012;79:1802-1808.
crossref pmid
397. Torres ER, Strack EF, Fernandez CE, Tumey TA, Hitchcock ME. Physical activity and white matter hyperintensities: a systematic review of quantitative studies. Prev Med Rep 2015;2:319-325.
crossref pmid pmc
398. Podewils LJ, Guallar E, Beauchamp N, Lyketsos CG, Kuller LH, Scheltens P. Physical activity and white matter lesion progression: assessment using MRI. Neurology 2007;68:1223-1226.
crossref pmid
399. Pan Y, Shen J, Cai X, Chen H, Zong G, Zhu W, et al. Adherence to a healthy lifestyle and brain structural imaging markers. Eur J Epidemiol 2023;38:657-668.
crossref pmid pdf


ABOUT JoS
AUTHOR INFORMATION
ARTICLE CATEGORY

Browse all articles >

BROWSE ARTICLES
Editorial Office
Department of Neurology, Asan Medical Center,Ulsan University College of Medicine
88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea
Submission, status and progress, etc ⟫ E-mail: editor@j-stroke.org
Website and system ⟫ E-mail: journal@m2community.co.kr
Publishing company ⟫ E-mail: ka72sus@smileml.com
Developed in M2PI
Copyright © 2024 by Korean Stroke Society.
Close layer
prev next